1
|
Jeeva P, Muthusamy A, Kesavan swaminathan J. Deciphering Structural Dynamics of Atherosclerosis Proteins: Insights from Crataegus oxyacantha Phytochemicals that Interceded Functional and Structural Changes in Targeted Atherosclerotic Proteins. ACS OMEGA 2024; 9:48159-48172. [PMID: 39676950 PMCID: PMC11635474 DOI: 10.1021/acsomega.4c04975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 12/17/2024]
Abstract
Atherosclerosis (ASC) is characterized by foam cell-mediated plaque formation, vascular endothelial inflammation, and lipidosis and is the rudimentary cause of cardiovascular diseases. This is the pre-eminent global factor of mortality. This etiological paradigm is significantly influenced by several proteins, where 23 pivotal proteins involved in ASC were meticulously gleaned on the basis of literature studies. The crux of the present study was aimed to probe the drugability of four active phytochemicals from Crataegus oxyacantha (COC): epicatechin, gallate, tyramine, and vitexin against the selected 23 proteins. The molecular docking analysis was judiciously administered via Glide, the binding free energy was calculated in detail utilizing the prime molecular mechanics-generalized Born surface area (MM-GBSA) module, and a deeper comprehensive investigation of protein-ligand dynamic associations was elucidated through Desmond. Drawing from the upper echelons of our docking results, the molecular dynamics simulation outcomes revealed that the macrophage migration inhibitory factor and prethrombin-1 showed persistent binding nature with gallate. The bioactive compound known as gallate sourced from COC shows the best molecular association with pivotal proteins involved in ASC and has a promising therapeutic potential for drug development endeavors.
Collapse
Affiliation(s)
- Praveen Jeeva
- Department
of Bioinformatics, Bharathidasan University, Tiruchirappalli, Tamilnadu 620024, India
| | - Anusuyadevi Muthusamy
- Department
of Biochemistry, Bharathidasan University, Tiruchirappalli, Tamilnadu 620024, India
| | | |
Collapse
|
2
|
Zhao L, Zhao BH, Ruze A, Li QL, Deng AX, Gao XM. Distinct roles of MIF in the pathogenesis of ischemic heart disease. Cytokine Growth Factor Rev 2024; 80:121-137. [PMID: 39438226 DOI: 10.1016/j.cytogfr.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
The role of macrophage migration inhibitory factor (MIF) as a multifunctional cytokine in immunomodulation and inflammatory response is increasingly appreciated. Ischemic heart disease (IHD), the leading cause of global mortality, remains a focal point of research owing to its intricate pathophysiology. MIF has been identified as a critical player in IHD, where it exerts distinct roles. On one hand, MIF plays a protective role by enhancing energy metabolism through activation of AMPK, resisting oxidative stress, inhibiting activation of the JNK pathway, and maintaining intracellular calcium ion homeostasis. Additionally, MIF exerts protective effects through mesenchymal stem cells and exosomes. On the other hand, MIF can assume a pro-inflammatory role, which contributes to the exacerbation of IHD's development and progression. Furthermore, MIF levels significantly increase in IHD patients, and its genetic polymorphisms are positively correlated with prevalence and severity. These findings position MIF as a potential biomarker and therapeutic target in the management of IHD. This review summarizes the structure, source, signaling pathways and biological functions of MIF and focuses on its roles and clinical characteristics in IHD. The genetic variants of MIF associated with IHD is also discussed, providing more understandings of its complex interplay in the disease's pathology.
Collapse
Affiliation(s)
- Ling Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Bang-Hao Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Amanguli Ruze
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Qiu-Lin Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - An-Xia Deng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Xiao-Ming Gao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China; Xinjiang Key Laboratory of Cardiovascular Disease, Urumqi, China.
| |
Collapse
|
3
|
Cheng YW, Yang LY, Chen YT, Chou SC, Chen KW, Chen YH, Deng CR, Chen IC, Chou WJ, Chang CC, Chen YR, Hwa HL, Wang KC, Kuo MF. Endothelial progenitor cell-derived conditioned medium mitigates chronic cerebral ischemic injury through macrophage migration inhibitory factor-activated AKT pathway. Stem Cell Res Ther 2024; 15:428. [PMID: 39543689 PMCID: PMC11566597 DOI: 10.1186/s13287-024-04015-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 10/26/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Chronic cerebral ischemia (CCI) is a significant health issue characterized by hypoperfusion due to damage or occlusion of the cerebral or carotid arteries. CCI may lead to progressive cognitive impairment that is considered as a prelude to neurodegenerative diseases, including dementia and Alzheimer's disease (AD). Endothelial progenitor cells (EPCs) have been implicated in vascular repair in ischemic cerebrovascular diseases, primarily by differentiating into endothelial cells (ECs) or through paracrine effects. However, the clinical transplantation of stem cell therapies remains limited. In this study, we investigated the effects of EPC-derived conditioned medium (EPC-CM) on the impaired vasculature and neurological function in a rodent model of CCI and the mechanism involved. METHODS EPC-CM was analyzed by cytokine array to identify key factors involved in angiogenesis and cellular senescence. The effects and mechanism of the candidate factors in the EPC-CM were validated in vitro using oxygen-glucose deprivation (OGD)-injured ECs and EPCs. The therapeutic effects of EPC-CM and the identified key factor were further examined in a rat model of CCI, which was induced by bilateral internal carotid artery ligation (BICAL). EPC-CM was administered via intracisternal injection one week post BICAL. The cerebral microvasculature and neurobehavior of the rats were examined three weeks after BICAL. RESULTS Macrophage migration inhibitory factor (MIF) was identified as a key factor in the EPC-CM. Recombinant MIF protein promoted angiogenesis and prevented senescence in the injured EPCs and ECs. The effect was similar to that of the EPC-CM. These therapeutic effects were diminished when the EPC-CM was co-treated with MIF-specific antibody (Ab). Additionally, the vascular, motor, and cognitive improvements observed in the BICAL rats treated with EPC-CM were abolished by co-treated with MIF Ab. Furthermore, we found MIF promoted angiogenesis and anti-senescence via activating the AKT pathway. Inhibition of the AKT pathway diminished the protective effects of MIF in the in vitro study. CONCLUSIONS We demonstrated that EPC-CM protected the brain from chronic ischemic injury and promoted functional recovery through MIF-mediated AKT pathway. These findings suggest EPC-CM holds potential as a novel cell-free therapeutic approach for treating CCI through the actions of MIF.
Collapse
Affiliation(s)
- Ya-Wen Cheng
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan
| | - Ling-Yu Yang
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan
| | - Yi-Tzu Chen
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Sheng-Che Chou
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Kuo-Wei Chen
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yi-Hsing Chen
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chuan-Rou Deng
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan
| | - I-Chin Chen
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan
| | - Wan-Ju Chou
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chen-Chih Chang
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yong-Ren Chen
- Non-Invasive Cancer Therapy Research Institute, Taipei, Taiwan
- Adjunct Visiting Staff, Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsiao-Lin Hwa
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
| | - Kuo-Chuan Wang
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan.
| | - Meng-Fai Kuo
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan.
| |
Collapse
|
4
|
Putha L, Kok LK, Fellner M, Rutledge MT, Gamble AB, Wilbanks SM, Vernall AJ, Tyndall JDA. Covalent Isothiocyanate Inhibitors of Macrophage Migration Inhibitory Factor as Potential Colorectal Cancer Treatments. ChemMedChem 2024; 19:e202400394. [PMID: 38977403 DOI: 10.1002/cmdc.202400394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/10/2024]
Abstract
Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine that has roles in innate and adaptive human immune responses, as well as inflammation. MIF exerts its biological activity by binding to the cell surface receptor CD74 as well as intracellular signalling proteins. MIF also possesses keto-enol tautomerase activity. Inhibition of the tautomerase activity has been associated with loss of biological activity of MIF and a potential anticancer target. Isothiocyanates (ITCs) are a class of compounds present in cruciferous vegetables that inhibit the MIF tautomerase activity via covalent modification of the N-terminal proline. A range of substituted ITCs featuring benzyl, phenethyl and phenyl propyl isothiocyanates were designed, synthesised and tested to determine any structure activity relationship for inhibiting MIF. Crystal structures of covalent compounds 8 and 9 in complex with rhMIF revealed key hydrogen bonding and edge-to-face π stacking interactions. Compound 9 and 11 with sub micromolar activity were tested in the NCI60 cancer cell lines panel. Both compounds showed tissue-specific reduced growth in colon and renal cancer cell lines, while one of these showed potent, dose-dependent inhibition of growth against all seven colon cancer cell lines (GI50<2.5 μM) and all eight renal cancer cell lines (GI50<2.2 μM).
Collapse
Affiliation(s)
- Lohitha Putha
- School of Pharmacy, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Liang K Kok
- School of Pharmacy, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Matthias Fellner
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Malcolm T Rutledge
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Allan B Gamble
- School of Pharmacy, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Sigurd M Wilbanks
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Andrea J Vernall
- Department of Chemistry, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Joel D A Tyndall
- School of Pharmacy, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| |
Collapse
|
5
|
Christopoulou ME, Aletras AJ, Papakonstantinou E, Stolz D, Skandalis SS. WISP1 and Macrophage Migration Inhibitory Factor in Respiratory Inflammation: Novel Insights and Therapeutic Potentials for Asthma and COPD. Int J Mol Sci 2024; 25:10049. [PMID: 39337534 PMCID: PMC11432718 DOI: 10.3390/ijms251810049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Recent advancements highlight the intricate interplay between the extracellular matrix (ECM) and immune responses, notably in respiratory diseases such as asthma and Chronic Obstructive Pulmonary Disease (COPD). The ECM, a dynamic structural framework within tissues, orches-trates a plethora of cellular processes, including immune cell behavior and tissue repair mecha-nisms. WNT1-inducible-signaling pathway protein 1 (WISP1), a key ECM regulator, controls immune cell behavior, cytokine production, and tissue repair by modulating integrins, PI3K, Akt, β-catenin, and mTOR signaling pathways. WISP1 also induces macrophage migration inhibitory factor (MIF) expression via Src kinases and epidermal growth factor receptor (EGFR) activation. MIF, through its wide range of activities, enhances inflammation and tissue restructuring. Rec-ognized for its versatile roles in regulating the immune system, MIF interacts with multiple immune components, such as the NLRP3 inflammasome, thereby sustaining inflammatory pro-cesses. The WISP1-MIF axis potentially unveils complex molecular mechanisms governing im-mune responses and inflammation. Understanding the intricate roles of WISP1 and MIF in the pathogenesis of chronic respiratory diseases such as asthma and COPD could lead to the identi-fication of novel targets for therapeutic intervention to alleviate disease severity and enhance patient outcomes.
Collapse
Affiliation(s)
- Maria-Elpida Christopoulou
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Alexios J Aletras
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Eleni Papakonstantinou
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Daiana Stolz
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Spyros S Skandalis
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
6
|
Khezrian A, Shojaeian A, Khaghani Boroujeni A, Amini R. Therapeutic Opportunities in Breast Cancer by Targeting Macrophage Migration Inhibitory Factor as a Pleiotropic Cytokine. Breast Cancer (Auckl) 2024; 18:11782234241276310. [PMID: 39246383 PMCID: PMC11380135 DOI: 10.1177/11782234241276310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/28/2024] [Indexed: 09/10/2024] Open
Abstract
As a heterogeneous disease, breast cancer (BC) has been characterized by the uncontrolled proliferation of mammary epithelial cells. The tumor microenvironment (TME) also contains inflammatory cells, fibroblasts, the extracellular matrix (ECM), and soluble factors that all promote BC progression. In this sense, the macrophage migration inhibitory factor (MIF), a pleiotropic pro-inflammatory cytokine and an upstream regulator of the immune response, enhances breast tumorigenesis through escalating cancer cell proliferation, survival, angiogenesis, invasion, metastasis, and stemness, which then brings tumorigenic effects by activating key oncogenic signaling pathways and inducing immunosuppression. Against this background, this review was to summarize the current understanding of the MIF pathogenic mechanisms in cancer, particularly BC, and address the central role of this immunoregulatory cytokine in signaling pathways and breast tumorigenesis. Furthermore, different inhibitors, such as small molecules as well as antibodies (Abs) or small interfering RNA (siRNA) and their anti-tumor effects in BC studies were examined. Small molecules and other therapy target MIF. Considering MIF as a promising therapeutic target, further clinical evaluation of MIF-targeted agents in patients with BC was warranted.
Collapse
Affiliation(s)
- Ali Khezrian
- Research Center for Molecular Medicine, Institute of Cancer, Avicenna Health Research Institute (AHRI), Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Shojaeian
- Research Center for Molecular Medicine, Institute of Cancer, Avicenna Health Research Institute (AHRI), Hamadan University of Medical Sciences, Hamadan, Iran
| | - Armin Khaghani Boroujeni
- Skin Disease and Leishmaniasis Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Razieh Amini
- Research Center for Molecular Medicine, Institute of Cancer, Avicenna Health Research Institute (AHRI), Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
7
|
Valdez CN, Sánchez-Zuno GA, Osmani L, Ibrahim W, Galan A, Bacchiocchi A, Halaban R, Kulkarni RP, Kang I, Bucala R, Tran T. Prognostic and therapeutic insights into MIF, DDT, and CD74 in melanoma. Oncotarget 2024; 15:507-520. [PMID: 39028303 PMCID: PMC11259151 DOI: 10.18632/oncotarget.28615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024] Open
Abstract
Macrophage Migration Inhibitory Factor (MIF) and its homolog D-dopachrome Tautomerase (DDT) have been implicated as drivers of tumor progression across a variety of cancers. Recent evidence suggests MIF as a therapeutic target in immune checkpoint inhibition (ICI) resistant melanomas, however clinical evidence of MIF and particularly of DDT remain limited. This retrospective study analyzed 97 patients treated at Yale for melanoma between 2002-2020. Bulk-RNA sequencing of patient tumor samples from the Skin Cancer SPORE Biorepository was used to evaluate for differential gene expression of MIF, DDT, CD74, and selected inflammatory markers, and gene expression was correlated with patient survival outcomes. Our findings revealed a strong correlation between MIF and DDT levels, with no statistically significant difference across common melanoma mutations and subtypes. Improved survival was associated with lower MIF and DDT levels and higher CD74:MIF and CD74:DDT levels. High CD74:DDT and CD74:MIF levels were also associated with enrichment of infiltrating inflammatory cell markers. These data suggest DDT as a novel target in immune therapy. Dual MIF and DDT blockade may provide synergistic responses in patients with melanoma, irrespective of common mutations, and may overcome ICI resistance. These markers may also provide prognostic value for further biomarker development.
Collapse
Affiliation(s)
| | | | - Lais Osmani
- Department of Medicine, Section of Rheumatology, Allergy and Immunology, Yale University, New Haven, CT 06520, USA
| | - Wael Ibrahim
- Department of Dermatology, Yale University, New Haven, CT 06520, USA
| | - Anjela Galan
- Department of Dermatology, Yale University, New Haven, CT 06520, USA
| | | | - Ruth Halaban
- Department of Dermatology, Yale University, New Haven, CT 06520, USA
| | - Rajan P. Kulkarni
- Department of Dermatology, Oregon Health and Science University, Portland, OR 97239, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
- Department of Veterans Affairs Portland Health Care System, Operative Care Division, U.S. Portland, OR 97239, USA
| | - Insoo Kang
- School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Medicine, Section of Rheumatology, Allergy and Immunology, Yale University, New Haven, CT 06520, USA
| | - Richard Bucala
- School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Medicine, Section of Rheumatology, Allergy and Immunology, Yale University, New Haven, CT 06520, USA
- Yale Cancer Center, Yale University, New Haven, CT 06520, USA
| | - Thuy Tran
- School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Medicine, Section of Medical Oncology, Yale University, New Haven, CT 06520, USA
- Yale Cancer Center, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
8
|
Wu KZ, Le Z, Myint B, Chan B, Liu L, Huang H, Sing SL, Tay A. Bioactive coating provides antimicrobial protection through immunomodulation and phage therapeutics. Mater Today Bio 2024; 26:101022. [PMID: 38525309 PMCID: PMC10959705 DOI: 10.1016/j.mtbio.2024.101022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/07/2024] [Accepted: 03/09/2024] [Indexed: 03/26/2024] Open
Abstract
Medical implant-associated infections (IAI) is a growing threat to patients undergoing implantation surgery. IAI prevention typically relies on medical implants endowed with bactericidal properties achieved through surface modifications with antibiotics. However, the clinical efficacy of this traditional paradigm remains suboptimal, often necessitating revision surgery and posing potentially lethal consequences for patients. To bolster the existing anti-IAI arsenal, we propose herein a chitosan-based bioactive coating, i.e., ChitoAntibac, which exerts bacteria-inhibitory effects either through immune modulation or phage-directed microbial clearance, without relying on conventional antibiotics. The immuno-stimulating effects and phage-induced bactericidal properties can be tailored by engineering the loading dynamic of macrophage migration inhibitory factor (MIF), which polarizes macrophages towards the proinflammatory subtype (M1) with enhanced bacterial phagocytosis, and Staphylococcal Phage K, resulting in rapid and targeted pathogenic clearance (>99.99%) in less than 8 h. Our innovative antibacterial coating opens a new avenue in the pursuit of effective IAI prevention through immuno-stimulation and phage therapeutics.
Collapse
Affiliation(s)
- Kenny Zhuoran Wu
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 119276, Singapore
| | - Zhicheng Le
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 119276, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Ba Myint
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 119276, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Brian Chan
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 119276, Singapore
| | - Ling Liu
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Hua Huang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore
- Electrophysiology Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore: Level 5, Centre for Life Sciences, 28 Medical Drive, 117456, Singapore
| | - Swee Leong Sing
- NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
- Department of Mechanical Engineering, College of Design and Engineering, National University of Singapore, 117575, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 119276, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
- NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
| |
Collapse
|
9
|
Tanese K, Ogata D. The role of macrophage migration inhibitory factor family and CD74 in the pathogenesis of melanoma. Exp Dermatol 2024; 33:e15122. [PMID: 38884501 DOI: 10.1111/exd.15122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 06/18/2024]
Abstract
Melanoma is an aggressive tumour with poor prognosis that arises from the malignant transformation of melanocytes. Over the past few decades, intense research into the pathogenesis of melanoma has led to the development of BRAF and immune checkpoint inhibitors, including antibodies against programmed cell death protein 1 (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4), which have shown clinically significant efficacy. However, some tumours do not respond to these therapies initially or become treatment resistant. Most melanoma tissues appear to possess biological characteristics that allow them to evade these treatments, and identifying these characteristics is one of the major challenges facing cancer researchers. One such characteristic that has recently gained attention is the role of macrophage migration inhibitory factor (MIF) and its receptor CD74. This review outlines the cellular and molecular functions of CD74, MIF and their family of proteins. We then review their roles in tumours based on previous reports, highlight their pathological significance in melanoma and discuss their potential as therapeutic targets.
Collapse
Affiliation(s)
- Keiji Tanese
- Department of Dermatology, Toho University School of Medicine, Tokyo, Japan
| | - Dai Ogata
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
10
|
Ye S, Agalave NM, Ma F, Mahmood DFD, Al-Grety A, Khoonsari PE, Leng L, Svensson CI, Bucala R, Kultima K, Vera PL. MIF-Modulated Spinal Proteins Associated with Persistent Bladder Pain: A Proteomics Study. Int J Mol Sci 2024; 25:4484. [PMID: 38674069 PMCID: PMC11050327 DOI: 10.3390/ijms25084484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Bladder pain is a prominent symptom in Interstitial Cystitis/Bladder Pain Syndrome (IC/BPS). We studied spinal mechanisms of bladder pain in mice using a model where repeated activation of intravesical Protease Activated Receptor-4 (PAR4) results in persistent bladder hyperalgesia (BHA) with little or no bladder inflammation. Persistent BHA is mediated by spinal macrophage migration inhibitory factor (MIF), and is associated with changes in lumbosacral proteomics. We investigated the contribution of individual spinal MIF receptors to persistent bladder pain as well as the spinal proteomics changes associated with relief of persistent BHA by spinal MIF antagonism. Female mice with persistent BHA received either intrathecal (i.t.) MIF monoclonal antibodies (mAb) or mouse IgG1 (isotype control antibody). MIF antagonism temporarily reversed persistent BHA (peak effect: 2 h), while control IgG1 had no effect. Moreover, i.t. antagonism of the MIF receptors CD74 and C-X-C chemokine receptor type 4 (CXCR4) partially reversed persistent BHA. For proteomics experiments, four separate groups of mice received either repeated intravesical scrambled peptide and sham i.t. injection (control, no pain group) or repeated intravesical PAR4 and: sham i.t.; isotype IgG1 i.t. (15 μg); or MIF mAb (15 μg). L6-S1 spinal segments were excised 2 h post-injection and examined for proteomics changes using LC-MS/MS. Unbiased proteomics analysis identified and relatively quantified 6739 proteins. We selected proteins that showed significant changes compared to control (no pain group) after intravesical PAR4 (sham or IgG i.t. treatment) and showed no significant change after i.t. MIF antagonism. Six proteins decreased during persistent BHA (V-set transmembrane domain-containing protein 2-like confirmed by immunohistochemistry), while two proteins increased. Spinal MIF antagonism reversed protein changes. Therefore, spinal MIF and MIF receptors mediate persistent BHA and changes in specific spinal proteins. These novel MIF-modulated spinal proteins represent possible new targets to disrupt spinal mechanisms that mediate persistent bladder pain.
Collapse
Affiliation(s)
- Shaojing Ye
- Research & Development, Lexington VA Health Care System, Lexington, KY 40502, USA; (S.Y.); (F.M.); (D.F.D.M.)
| | - Nilesh M. Agalave
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, SE-751 85 Uppsala, Sweden; (N.M.A.); (A.A.-G.); (P.E.K.); (K.K.)
| | - Fei Ma
- Research & Development, Lexington VA Health Care System, Lexington, KY 40502, USA; (S.Y.); (F.M.); (D.F.D.M.)
| | - Dlovan F. D. Mahmood
- Research & Development, Lexington VA Health Care System, Lexington, KY 40502, USA; (S.Y.); (F.M.); (D.F.D.M.)
| | - Asma Al-Grety
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, SE-751 85 Uppsala, Sweden; (N.M.A.); (A.A.-G.); (P.E.K.); (K.K.)
| | - Payam E. Khoonsari
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, SE-751 85 Uppsala, Sweden; (N.M.A.); (A.A.-G.); (P.E.K.); (K.K.)
| | - Lin Leng
- Department of Internal Medicine, Yale University, New Haven, CT 06510, USA; (L.L.); (R.B.)
| | - Camilla I. Svensson
- Department of Physiology and Pharmacology, Karolinska Institutet (KI), SE-171 65 Solna, Sweden;
| | - Richard Bucala
- Department of Internal Medicine, Yale University, New Haven, CT 06510, USA; (L.L.); (R.B.)
| | - Kim Kultima
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, SE-751 85 Uppsala, Sweden; (N.M.A.); (A.A.-G.); (P.E.K.); (K.K.)
| | - Pedro L. Vera
- Research & Development, Lexington VA Health Care System, Lexington, KY 40502, USA; (S.Y.); (F.M.); (D.F.D.M.)
- Department of Physiology, University of Kentucky, Lexington, KY 40506, USA
| |
Collapse
|
11
|
Souza Silva VR, Mota CM, Maia LP, Ferreira FB, Miranda VDS, Silva NM, Ferro EAV, Mineo JR, Mineo TWP. Macrophage migration inhibitory factor favors Neospora caninum infection in mice. Microb Pathog 2024; 189:106577. [PMID: 38367848 DOI: 10.1016/j.micpath.2024.106577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 02/06/2024] [Accepted: 02/15/2024] [Indexed: 02/19/2024]
Abstract
Neospora caninum is a protozoan parasite with worldwide incidence, acting as a major cause of reproductive failures in ruminants and neuromuscular symptoms in dogs. Macrophage Migration Inhibitory Factor (MIF) is produced by several cell types and exhibits a central role in immune responses against intracellular pathogens. The present study aimed to comprehend the role of MIF in the relationship between N. caninum and its host. We used in vivo, in vitro and ex vivo experiments in a model of infection based on genetically deficient mice to analyze the infection kinetics and inflammatory markers. MIF production was measured in response to N. caninum during the acute and chronic phases of the infection. While Mif-/- mice survived lethal doses of NcLiv tachyzoites, sublethal infections in these mice showed that parasite burden was controlled in target tissues, alongside with reduced inflammatory infiltrates detected in lung and brain sections. TNF was increased at the initial site of the infection in genetically deficient mice and the MIF-dependent reduction was confirmed in vitro with macrophages and ex vivo with primed spleen cells. In sum, MIF negatively regulated host immunity against N. caninum, favoring disease progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Eloísa A Vieira Ferro
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia. Av. Amazonas, Campus Umuarama., 38405-320, Uberlândia, Minas Gerais, Brazil
| | | | | |
Collapse
|
12
|
Silva RCMC, Gomes FM. Evolution of the Major Components of Innate Immunity in Animals. J Mol Evol 2024; 92:3-20. [PMID: 38281163 DOI: 10.1007/s00239-024-10155-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 01/11/2024] [Indexed: 01/30/2024]
Abstract
Innate immunity is present in all animals. In this review, we explore the main conserved mechanisms of recognition and innate immune responses among animals. In this sense, we discuss the receptors, critical for binding to pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs); the downstream signaling proteins; and transcription factors that govern immune responses. We also highlight conserved inflammatory mediators that are induced after the recognition of DAMPs and PAMPs. At last, we discuss the mechanisms that are involved in the regulation and/or generation of reactive oxygen species (ROS), influencing immune responses, like heme-oxygenases (HOs).
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratory of Immunoreceptors and Signaling, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Fábio Mendonça Gomes
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
13
|
Talreja J, Peng C, Samavati L. MIF modulates p38/ERK phosphorylation via MKP-1 induction in sarcoidosis. iScience 2024; 27:108746. [PMID: 38299032 PMCID: PMC10829885 DOI: 10.1016/j.isci.2023.108746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 09/21/2023] [Accepted: 12/12/2023] [Indexed: 02/02/2024] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a versatile cytokine that influences a variety of cellular processes important for immune regulation and tissue homeostasis. Sarcoidosis is a granulomatous disease characterized by extensive local inflammation and increased T helper cell mediated cytokines. We have shown that MIF has a modulatory role in cytokine networks in sarcoidosis. We investigated the effect of exogenous MIF on sarcoidosis alveolar macrophages (AMs), CD14+ monocytes and peripheral blood mononuclear cells (PBMCs). Our results showed that MIF negatively regulates the increased MAPKs (pp38 and pERK1/2) activation by inducing Mitogen-activated protein kinase phosphatase (MKP)-1. We found that MIF decreased IL-6 and IL-1β production, increased the percentage of regulatory T-cells (Tregs), and induced IL-1R antagonist (IL-1RA) and IL-10 production. Thus, the results of our study suggest that exogenous MIF modulates MAPK activation by inducing MKP-1and Tregs as well as IL-10 and IL-1RA, and hence plays a modulatory role in immune activation in sarcoidosis.
Collapse
Affiliation(s)
- Jaya Talreja
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University, School of Medicine and Detroit Medical Center, Detroit, MI 48201, USA
| | - Changya Peng
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University, School of Medicine and Detroit Medical Center, Detroit, MI 48201, USA
| | - Lobelia Samavati
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University, School of Medicine and Detroit Medical Center, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI 48201, USA
| |
Collapse
|
14
|
Matejuk A, Benedek G, Bucala R, Matejuk S, Offner H, Vandenbark AA. MIF contribution to progressive brain diseases. J Neuroinflammation 2024; 21:8. [PMID: 38178143 PMCID: PMC10765708 DOI: 10.1186/s12974-023-02993-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 12/12/2023] [Indexed: 01/06/2024] Open
Abstract
Progressive brain diseases create a huge social and economic burden on modern societies as a major cause of disability and death. Incidence of brain diseases has a significantly increasing trend and merits new therapeutic strategies. At the base of many progressive brain malfunctions is a process of unresolved, chronic inflammation. Macrophage migration inhibitory factor, MIF, is an inflammatory mediator that recently gained interest of neuro-researchers due to its varied effects on the CNS such as participation of nervous system development, neuroendocrine functions, and modulation of neuroinflammation. MIF appears to be a candidate as a new biomarker and target of novel therapeutics against numerous neurologic diseases ranging from cancer, autoimmune diseases, vascular diseases, neurodegenerative pathology to psychiatric disorders. In this review, we will focus on MIF's crucial role in neurological diseases such as multiple sclerosis (MS), Alzheimer's disease (AD) and glioblastoma (GBM).
Collapse
Affiliation(s)
- Agata Matejuk
- Department of Immunology, Collegium Medicum, University of Zielona Góra, Zielona Góra, Poland.
| | - Gil Benedek
- Tissue Typing and Immunogenetics Unit, Department of Genetics, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Richard Bucala
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | | | - Halina Offner
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA
- Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - Arthur A Vandenbark
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA.
- Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
| |
Collapse
|
15
|
Ladakis DC, Reyes-Mantilla MI, Gadani SP, Mace JW, Dominguez-Penuela SC, Appiah MJ, Smith MD, Bhargava P, Fox RJ, Saidha S, Calabresi PA. Serum macrophage migration inhibitory factor levels predict brain atrophy in people with primary progressive multiple sclerosis. Mult Scler 2024; 30:35-43. [PMID: 37982154 DOI: 10.1177/13524585231213164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF) is a cytokine linked to multiple sclerosis (MS) progression that is thought to be inhibited by ibudilast. SPRINT-MS was a phase 2 placebo-controlled trial of ibudilast in progressive multiple sclerosis (PMS). OBJECTIVE To determine whether baseline MIF levels predict imaging outcomes and assess the effects of ibudilast on serum and cerebrospinal fluid (CSF) MIF levels in people with PMS treated with ibudilast. METHODS Participants in the SPRINT-MS trial were treated with either ibudilast or placebo and underwent brain magnetic resonance imaging (MRI) every 24 weeks over a duration of 96 weeks. MIF was measured in serum and CSF. RESULTS MIF levels were compared with imaging outcomes in 223 participants from the SPRINT-MS study. In the primary progressive multiple sclerosis (PPMS) cohort, males had higher serum (p < 0.001) and CSF (p = 0.01) MIF levels, as compared with females. Higher baseline serum MIF levels in PPMS were associated with faster brain atrophy (beta = -0.113%, 95% confidence interval (CI): -0.204% to -0.021%; p = 0.016). These findings were not observed in secondary progressive multiple sclerosis (SPMS). Ibudilast did not affect either serum or CSF MIF levels. CONCLUSIONS Serum MIF levels were associated with male sex and predicted brain atrophy in PPMS, but not SPMS. Ibudilast did not demonstrate an effect on MIF levels, as compared with placebo, although we cannot exclude a functional effect.
Collapse
Affiliation(s)
- Dimitrios C Ladakis
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maria I Reyes-Mantilla
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sachin P Gadani
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jackson W Mace
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Mayaa J Appiah
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew D Smith
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pavan Bhargava
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert J Fox
- Mellen Center for Multiple Sclerosis, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Shiv Saidha
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Su Y, Zhang X, Liang Y, Sun J, Lu C, Huang Z. Integrated analysis of single-cell RNA-seq and bulk RNA-seq to unravel the molecular mechanisms underlying the immune microenvironment in the development of intestinal-type gastric cancer. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166849. [PMID: 37591405 DOI: 10.1016/j.bbadis.2023.166849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/02/2023] [Accepted: 08/12/2023] [Indexed: 08/19/2023]
Abstract
Intestinal-type gastric cancer (IGC) is the most frequent type of gastric cancer in high-incidence populations. The early stages of IGC growth successively include nonatrophic gastritis (NAG), chronic atrophic gastritis (CAG) and intestinal metaplasia (IM). However, the mechanisms of IGC development through these stages remain unclear. For this study, single-cell RNA-seq data related to IGC were downloaded from the GEO database, and immune cells of the tumor microenvironment (TME) were annotated using R software. Changes in the proportion of immune cells and altered cell-to-cell interactions were explored at different disease stages using R software, with a focus on plasma cells. Additionally, IGC samples from the TCGA database were used for immune cell infiltration analysis, and a Cox proportional risk regression model was constructed to identify possible prognostic genes. The results indicated that for precancerous lesions, interactions between immune cells were mainly dominated by chemokines to stimulate the infiltration and activation of immune cells. In tumors, intercellular movement of upregulated molecules and amplified signals were associated with the tumor necrosis factor family and immunosuppression to escape immune surveillance and promote tumor growth. Regarding prognostic analysis, IGLC3, IGLV1-44, IGKV1-16, IGHV3-21, IGLV1-51, and IGLV3-19 were found to be novel biomarkers for IGC. Our analysis of the IGC single-cell atlas together with bulk transcriptome data contributes to understanding TME heterogeneity at the molecular level during IGC development and provides insights for elucidating the mechanism of IGC and discovering novel targets for precise therapy.
Collapse
Affiliation(s)
- Yongjian Su
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China; Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Xiaoqing Zhang
- School of Basic Medicine, Guangdong Medical University, Dongguan, China
| | - Youcheng Liang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China; Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Jianbo Sun
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Chengyu Lu
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, China.
| | - Zunnan Huang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China; Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, China.
| |
Collapse
|
17
|
Breidung D, Megas IF, Freytag DL, Bernhagen J, Grieb G. The Role of Macrophage Migration Inhibitory Factor (MIF) and D-Dopachrome Tautomerase (D-DT/MIF-2) in Infections: A Clinical Perspective. Biomedicines 2023; 12:2. [PMID: 38275363 PMCID: PMC10813530 DOI: 10.3390/biomedicines12010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
Macrophage migration inhibitory factor (MIF) and its homolog, D-dopachrome tautomerase (D-DT), are cytokines that play critical roles in the immune response to various infectious diseases. This review provides an overview of the complex involvement of MIF and D-DT in bacterial, viral, fungal, and parasitic infections. The role of MIF in different types of infections is controversial, as it has either a protective function or a host damage-enhancing function depending on the pathogen. Depending on the specific role of MIF, different therapeutic options for MIF-targeting drugs arise. Human MIF-neutralizing antibodies, anti-parasite MIF antibodies, small molecule MIF inhibitors or MIF-blocking peptides, as well as the administration of exogenous MIF or MIF activity-augmenting small molecules have potential therapeutic applications and need to be further explored in the future. In addition, MIF has been shown to be a potential biomarker and therapeutic target in sepsis. Further research is needed to unravel the complexity of MIF and D-DT in infectious diseases and to develop personalized therapeutic approaches targeting these cytokines. Overall, a comprehensive understanding of the role of MIF and D-DT in infections could lead to new strategies for the diagnosis, treatment, and management of infectious diseases.
Collapse
Affiliation(s)
- David Breidung
- Department of Plastic, Reconstructive and Hand Surgery, Burn Center for Severe Burn Injuries, Klinikum Nuremberg Hospital, Paracelsus Medical University, Breslauer Str. 201, 90471 Nuremberg, Germany;
| | - Ioannis-Fivos Megas
- Department of Orthopaedic and Trauma Surgery, Center of Plastic Surgery, Hand Surgery and Microsurgery, Evangelisches Waldkrankenhaus Spandau, Stadtrandstr. 555, 13589 Berlin, Germany;
| | - David Lysander Freytag
- Department of Plastic Surgery and Hand Surgery, Gemeinschaftskrankenhaus Havelhoehe, Kladower Damm 221, 14089 Berlin, Germany;
| | - Jürgen Bernhagen
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München (KUM), Ludwig-Maximilians-University (LMU), Feodor-Lynenstraße 17, 81377 Munich, Germany;
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynenstraße 17, 81377 Munich, Germany
| | - Gerrit Grieb
- Department of Plastic Surgery and Hand Surgery, Gemeinschaftskrankenhaus Havelhoehe, Kladower Damm 221, 14089 Berlin, Germany;
- Department of Plastic Surgery and Hand Surgery, Burn Center, Medical Faculty, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany
| |
Collapse
|
18
|
Wang Y, Dang Z, Wang X, Chen Y, Dong P, Liu G, Tan W, Gui Z, Bu F, Lin F, Liang C. Obacunone alleviates chronic pelvic pain and pro-inflammatory depolarization of macrophage induced by experimental autoimmune prostatitis in mice. Biochem Biophys Rep 2023; 36:101565. [PMID: 37965064 PMCID: PMC10641089 DOI: 10.1016/j.bbrep.2023.101565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/09/2023] [Accepted: 10/23/2023] [Indexed: 11/16/2023] Open
Abstract
Chronic pelvic pain syndrome (CPPS) is a common complication of prostatitis, which was associated with the pathological depolarization of macrophage and the neuroinflammation. However, its underlying reason is far from clear and few effective treatments is applicable. In this study, we tested the effect of obacunone (Oba), a highly oxygenated triterpenoid, on CPPS. The experimental autoimmune prostatitis (EAP) was induced by subcutaneous injection of heterologous prostate homogenate in mice. We found that EAP led to prostatodynia, neuronal activation of spinal dorsal horn, and the pro-inflammatory depolarization of macrophage within prostate, which was significantly alleviated by oral administration of Oba in a dose-dependent manner. Mechanistically, EAP-induced production of IL-6 on prostatic macrophage was suppressed by Oba. Moreover, co-administration of Oba and MIF inhibitor ISO-1 did not lead to additive effect when compared with either alone. In summary, we conclude that Oba prevents the production of macrophage-derived pro-inflammatory factors by inhibiting MIF, which eventually alleviates CPPS after prostatitis.
Collapse
Affiliation(s)
- Yadong Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
- Department of Urology, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Zhaohui Dang
- Department of Neurology & Psychology, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Xu Wang
- Department of Neurology & Psychology, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Yuanyuan Chen
- Department of Neurology & Psychology, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Peng Dong
- Department of Neurosurgery, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Gang Liu
- Department of Neurosurgery, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Weibin Tan
- Department of Neurosurgery, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Zhong Gui
- Department of Neurosurgery, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Fan Bu
- Department of Neurology & Psychology, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Feng Lin
- Department of Urology, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| |
Collapse
|
19
|
Hao H, Hou Y, Li A, Niu L, Li S, He B, Zhang X, Song H, Cai R, Zhou Y, Yao C, Wang Y, Wang Y. HIF-1α promotes astrocytic production of macrophage migration inhibitory factor following spinal cord injury. CNS Neurosci Ther 2023; 29:3802-3814. [PMID: 37334735 PMCID: PMC10651974 DOI: 10.1111/cns.14300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/16/2023] [Accepted: 05/28/2023] [Indexed: 06/20/2023] Open
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF) is an important mediator of neuropathology in various central nervous system (CNS) diseases. However, little is known about its inducers for production from the nerve cells, as well as the underlying regulatory mechanism. Injury-induced HIF-1α has been shown to exacerbate neuroinflammation by activating multiple downstream target molecules. It is postulated that HIF-1α is involved in the regulation of MIF following spinal cord injury (SCI). METHODS SCI model of Sprague-Dawley rats was established by cord contusion at T8-T10. The dynamic changes of HIF-1α and MIF protein levels at lesion site of rat spinal cord were determined by Western blot. The specific cell types of HIF-1α and MIF expression were examined by immunostaining. Primary astrocytes were isolated from the spinal cord, cultured and stimulated with various agonist or inhibitor of HIF-1α for analysis of HIF-1α-mediated expression of MIF. Luciferase report assay was used to determine the relationship between HIF-1α and MIF. The Basso, Beattie, and Bresnahan (BBB) locomotor scale was used to assess the locomotor function following SCI. RESULTS The protein levels of HIF-1α and MIF at lesion site were significantly elevated by SCI. Immunofluorescence demonstrated that both HIF-1α and MIF were abundantly expressed in the astrocytes of the spinal cord. By using various agonists or inhibitors of HIF-1α, it was shown that HIF-1α sufficiently induced astrocytic production of MIF. Mechanistically, HIF-1α promoted MIF expression through interaction with MIF promoter. Inhibition of HIF-1α activity using specific inhibitor markedly reduced the protein levels of MIF at lesion site following SCI, which in turn favored for the functional recovery. CONCLUSION SCI-induced activation of HIF-1α is able to promote MIF production from astrocytes. Our results have provided new clues for SCI-induced production of DAMPs, which may be helpful for clinical treatment of neuroinflammation.
Collapse
Affiliation(s)
- Huifei Hao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Yuxuan Hou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Aicheng Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Li Niu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Shaolan Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Bingqiang He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Xingyuan Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Honghua Song
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Rixin Cai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Yue Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Chun Yao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Yongjun Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Yingjie Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| |
Collapse
|
20
|
Vámos E, Kálmán N, Sturm EM, Nayak BB, Teppan J, Vántus VB, Kovács D, Makszin L, Loránd T, Gallyas F, Radnai B. Highly Selective MIF Ketonase Inhibitor KRP-6 Diminishes M1 Macrophage Polarization and Metabolic Reprogramming. Antioxidants (Basel) 2023; 12:1790. [PMID: 37891870 PMCID: PMC10604361 DOI: 10.3390/antiox12101790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/14/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Macrophage polarization is highly involved in autoimmunity. M1 polarized macrophages drive inflammation and undergo metabolic reprogramming, involving downregulation of mitochondrial energy production and acceleration of glycolysis. Macrophage migration inhibitory factor (MIF), an enigmatic tautomerase (ketonase and enolase), was discovered to regulate M1 polarization. Here, we reveal that KRP-6, a potent and highly selective MIF ketonase inhibitor, reduces MIF-induced human blood eosinophil and neutrophil migration similarly to ISO-1, the most investigated tautomerase inhibitor. We equally discovered that KRP-6 prevents M1 macrophage polarization and reduces ROS production in IFN-γ-treated cells. During metabolic reprogramming, KRP-6 improved mitochondrial bioenergetics by ameliorating basal respiration, ATP production, coupling efficiency and maximal respiration in LPS+IFN-γ-treated cells. KRP-6 also reduced glycolytic flux in M1 macrophages. Moreover, the selective MIF ketonase inhibitor attenuated LPS+IFN-γ-induced downregulation of PARP-1 and PARP-2 mRNA expression. We conclude that KRP-6 represents a promising novel therapeutic compound for autoimmune diseases, which strongly involves M1 macrophage polarization.
Collapse
Affiliation(s)
- Eszter Vámos
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 12 Szigeti Str., 7624 Pécs, Hungary; (E.V.); (N.K.); (V.B.V.); (D.K.); (T.L.)
| | - Nikoletta Kálmán
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 12 Szigeti Str., 7624 Pécs, Hungary; (E.V.); (N.K.); (V.B.V.); (D.K.); (T.L.)
| | - Eva Maria Sturm
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria; (E.M.S.); (B.B.N.); (J.T.)
| | - Barsha Baisakhi Nayak
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria; (E.M.S.); (B.B.N.); (J.T.)
| | - Julia Teppan
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria; (E.M.S.); (B.B.N.); (J.T.)
| | - Viola Bagóné Vántus
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 12 Szigeti Str., 7624 Pécs, Hungary; (E.V.); (N.K.); (V.B.V.); (D.K.); (T.L.)
| | - Dominika Kovács
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 12 Szigeti Str., 7624 Pécs, Hungary; (E.V.); (N.K.); (V.B.V.); (D.K.); (T.L.)
| | - Lilla Makszin
- Institute of Bioanalysis, Medical School, Szentágothai Research Center, University of Pécs, 7622 Pécs, Hungary;
| | - Tamás Loránd
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 12 Szigeti Str., 7624 Pécs, Hungary; (E.V.); (N.K.); (V.B.V.); (D.K.); (T.L.)
| | - Ferenc Gallyas
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 12 Szigeti Str., 7624 Pécs, Hungary; (E.V.); (N.K.); (V.B.V.); (D.K.); (T.L.)
| | - Balázs Radnai
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 12 Szigeti Str., 7624 Pécs, Hungary; (E.V.); (N.K.); (V.B.V.); (D.K.); (T.L.)
| |
Collapse
|
21
|
Zhang H, Hu YM, Wang YJ, Zhou Y, Zhu ZJ, Chen MH, Wang YJ, Xu H, Wang YH. Macrophage migration inhibitory factor facilitates astrocytic production of the CCL2 chemokine following spinal cord injury. Neural Regen Res 2023; 18:1802-1808. [PMID: 36751809 PMCID: PMC10154479 DOI: 10.4103/1673-5374.363184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 10/08/2022] [Accepted: 10/20/2022] [Indexed: 12/14/2022] Open
Abstract
Spinal cord injury causes accumulation of a large number of leukocytes at the lesion site where they contribute to excessive inflammation. Overproduced chemokines are responsible for the migratory process of the leukocytes, but the regulatory mechanism underlying the production of chemokines from resident cells of the spinal cord has not been fully elucidated. We examined the protein levels of macrophage migration inhibitory factor and chemokine C-C motif chemokine ligand 2 in a spinal cord contusion model at different time points following spinal cord injury. The elevation of macrophage migration inhibitory factor at the lesion site coincided with the increase of chemokine C-C motif chemokine ligand 2 abundance in astrocytes. Stimulation of primary cultured astrocytes with different concentrations of macrophage migration inhibitory factor recombinant protein induced chemokine C-C motif chemokine ligand 2 production from the cells, and the macrophage migration inhibitory factor inhibitor 4-iodo-6-phenylpyrimidine attenuated the stimulatory effect. Further investigation into the underlying mechanism on macrophage migration inhibitory factor-mediated astrocytic production of chemokine C-C motif chemokine ligand 2 revealed that macrophage migration inhibitory factor activated intracellular JNK signaling through binding with CD74 receptor. Administration of the macrophage migration inhibitory factor inhibitor 4-iodo-6-phenylpyrimidine following spinal cord injury resulted in the reduction of chemokine C-C motif chemokine ligand 2-recruited microglia/macrophages at the lesion site and remarkably improved the hindlimb locomotor function of rats. Our results have provided insights into the functions of astrocyte-activated chemokines in the recruitment of leukocytes and may be beneficial to develop interventions targeting chemokine C-C motif chemokine ligand 2 for neuroinflammation after spinal cord injury.
Collapse
Affiliation(s)
- Han Zhang
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province, China
| | - Yu-Ming Hu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Ying-Jie Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Yue Zhou
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Zhen-Jie Zhu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Min-Hao Chen
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province, China
| | - Yong-Jun Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Hua Xu
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province, China
| | - You-Hua Wang
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
22
|
Zhang T, Luu MDA, Dolga AM, Eisel ULM, Schmidt M. The old second messenger cAMP teams up with novel cell death mechanisms: potential translational therapeutical benefit for Alzheimer's disease and Parkinson's disease. Front Physiol 2023; 14:1207280. [PMID: 37405135 PMCID: PMC10315612 DOI: 10.3389/fphys.2023.1207280] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/07/2023] [Indexed: 07/06/2023] Open
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) represent the most prevalent neurodegenerative disorders severely impacting life expectancy and quality of life of millions of people worldwide. AD and PD exhibit both a very distinct pathophysiological disease pattern. Intriguingly, recent researches, however, implicate that overlapping mechanisms may underlie AD and PD. In AD and PD, novel cell death mechanisms, encompassing parthanatos, netosis, lysosome-dependent cell death, senescence and ferroptosis, apparently rely on the production of reactive oxygen species, and seem to be modulated by the well-known, "old" second messenger cAMP. Signaling of cAMP via PKA and Epac promotes parthanatos and induces lysosomal cell death, while signaling of cAMP via PKA inhibits netosis and cellular senescence. Additionally, PKA protects against ferroptosis, whereas Epac1 promotes ferroptosis. Here we review the most recent insights into the overlapping mechanisms between AD and PD, with a special focus on cAMP signaling and the pharmacology of cAMP signaling pathways.
Collapse
Affiliation(s)
- Tong Zhang
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands
| | - Minh D. A. Luu
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | - Amalia M. Dolga
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | - Ulrich L. M. Eisel
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
23
|
Orlandi G, Roncucci L, Carnevale G, Sena P. Different Roles of Apoptosis and Autophagy in the Development of Human Colorectal Cancer. Int J Mol Sci 2023; 24:10201. [PMID: 37373349 DOI: 10.3390/ijms241210201] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Colorectal cancer (CRC) remains a major life-threatening malignancy, despite numerous therapeutic and screening attempts. Apoptosis and autophagy are two processes that share common signaling pathways, are linked by functional relationships and have similar protein components. During the development of cancer, the two processes can trigger simultaneously in the same cell, causing, in some cases, an inhibition of autophagy by apoptosis or apoptosis by autophagy. Malignant cells that have accumulated genetic alterations can take advantage of any alterations in the apoptotic process and as a result, progress easily in the cancerous transformation. Autophagy often plays a suppressive role during the initial stages of carcinogenicity, while in the later stages of cancer development it can play a promoting role. It is extremely important to determine the regulation of this duality of autophagy in the development of CRC and to identify the molecules involved, as well as the signals and the mechanisms behind it. All the reported experimental results indicate that, while the antagonistic effects of autophagy and apoptosis occur in an adverse environment characterized by deprivation of oxygen and nutrients, leading to the formation and development of CRC, the effects of promotion and collaboration usually involve an auxiliary role of autophagy compared to apoptosis. In this review, we elucidate the different roles of autophagy and apoptosis in human CRC development.
Collapse
Affiliation(s)
- Giulia Orlandi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Via del Pozzo, 71-41124 Modena, Italy
| | - Luca Roncucci
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Via del Pozzo, 71-41124 Modena, Italy
| | - Gianluca Carnevale
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Via del Pozzo, 71-41124 Modena, Italy
| | - Paola Sena
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Via del Pozzo, 71-41124 Modena, Italy
| |
Collapse
|
24
|
Liu W, Lin T, Gong L. ZD6474 Attenuates Fibrosis and Inhibits Neovascularization in Human Pterygium by Suppressing AKT-mTOR Signaling Pathway. J Ocul Pharmacol Ther 2023; 39:128-138. [PMID: 36576784 DOI: 10.1089/jop.2022.0127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Purpose: To investigate the antifibrotic effect of ZD6474 in human pterygium fibroblasts (HPFs) and angiogenesis in human umbilical vein endothelial cells (HUVECs) compared with mitomycin C (MMC). Methods: Pterygium and tenon fibroblasts were isolated from patients undergoing surgery to culture HPFs and human tenon fibroblasts (HTFs). The effects of ZD6474 on HPF, HTF, and HUVEC proliferation and migration were detected using CCK8 and wound-healing assays, respectively. Fibrosis and epithelial-mesenchymal transformation (EMT) were evaluated by western blotting [transforming growth factor beta (TGF-β)1/2 and snail] and immunofluorescence (vimentin and α-smooth muscle actin). The antiangiogenic effect of ZD6474 on HUVECs was assessed using a tube formation assay. To determine the potential mechanism, the expression of phosphorylated AKT (p-AKT) and phosphorylated mTOR (p-mTOR) was evaluated by treatment with ZD6474 via western blotting. Results: ZD6474 robustly inhibited the proliferation and migration of HPFs rather than HTFs compared with those in the MMC group (**P < 0.01). In HPFs, fibrosis and EMT (vimentin, TGF-β1/2, and snail) were significantly reversed by ZD6474. MMC (>50 μg/mL) significantly reduced HTF viability, whereas ZD6474 (<5 μM/mL) did not decrease HTF viability. HUVEC proliferation and migration were clearly decreased, and tube formation was notably interrupted by ZD6474. Activation of p-AKT and p-mTOR was inhibited by ZD6474 treatment of HPFs and HUVECs. Conclusion: ZD6474 is more effective than MMC in reducing fibrosis and EMT in HPFs. In addition, ZD6474 was less toxic to HTFs. ZD6474 also exhibited antiangiogenic effects in HUVECs. This study may aid in the development of novel agents to prevent pterygium recurrence after pterygium excision.
Collapse
Affiliation(s)
- Wenting Liu
- Department of Ophthalmology and Vision Science, The Eye, Ear, Nose and Throat Hospital of Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia, Laboratory of Myopia, Chinese Academy of Medical Sciences, Fudan University, Shanghai, China
- Department of Ophthalmology, Huadong Hospital of Fudan University, Shanghai, China
| | - Tong Lin
- Department of Ophthalmology and Vision Science, The Eye, Ear, Nose and Throat Hospital of Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia, Laboratory of Myopia, Chinese Academy of Medical Sciences, Fudan University, Shanghai, China
| | - Lan Gong
- Department of Ophthalmology and Vision Science, The Eye, Ear, Nose and Throat Hospital of Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia, Laboratory of Myopia, Chinese Academy of Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Wang L, Yang J, Tan W, Guo Y, Li J, Duan C, Wei G, Chou M. Macrophage migration inhibitory factor MtMIF3 prevents the premature aging of Medicago truncatula nodules. PLANT, CELL & ENVIRONMENT 2023; 46:1004-1017. [PMID: 36515398 DOI: 10.1111/pce.14515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 12/01/2022] [Accepted: 12/09/2022] [Indexed: 06/17/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine involved in immune response in animals. However, the role of MIFs in plants such as Medicago truncatula, particularly in symbiotic nitrogen fixation, remains unclear. An investigation of M. truncatula-Sinorhizobium meliloti symbiosis revealed that MtMIF3 was mainly expressed in the nitrogen-fixing zone of the nodules. Silencing MtMIF3 using RNA interference (Ri) technology resulted in increased nodule numbers but higher levels of bacteroid degradation in the infected cells of the nitrogen-fixing zone, suggesting that premature aging was induced in MtMIF3-Ri nodules. In agreement with this conclusion, the activities of nitrogenase, superoxide dismutase and catalase were lower than those in controls, but cysteine proteinase activity was increased in nodulated roots at 28 days postinoculation. In contrast, the overexpression of MtMIF3 inhibited nodule senescence. MtMIF3 is localized in the plasma membrane, nucleus, and cytoplasm, where it interacts with methionine sulfoxide reductase B (MsrB), which is also localized in the chloroplasts of tobacco leaf cells. Taken together, these results suggest that MtMIF3 prevents premature nodule aging and protects against oxidation by interacting with MtMsrB.
Collapse
Affiliation(s)
- Li Wang
- State Key Laboratory of Crop Stress Biology in Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Jieyu Yang
- State Key Laboratory of Crop Stress Biology in Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Wenjun Tan
- State Key Laboratory of Crop Stress Biology in Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Yile Guo
- State Key Laboratory of Crop Stress Biology in Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Jiaqi Li
- State Key Laboratory of Crop Stress Biology in Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Chuntao Duan
- State Key Laboratory of Crop Stress Biology in Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Gehong Wei
- State Key Laboratory of Crop Stress Biology in Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Minxia Chou
- State Key Laboratory of Crop Stress Biology in Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| |
Collapse
|
26
|
Hallmarks of Cancer Affected by the MIF Cytokine Family. Cancers (Basel) 2023; 15:cancers15020395. [PMID: 36672343 PMCID: PMC9856758 DOI: 10.3390/cancers15020395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
New diagnostic methods and treatments have significantly decreased the mortality rates of cancer patients, but further improvements are warranted based on the identification of novel tumor-promoting molecules that can serve as therapeutic targets. The macrophage migration inhibitory factor (MIF) family of cytokines, comprising MIF and DDT (also known as MIF2), are overexpressed in almost all cancer types, and their high expressions are related to a worse prognosis for the patients. MIF is involved in 9 of the 10 hallmarks of cancer, and its inhibition by antibodies, nanobodies, or small synthetic molecules has shown promising results. Even though DDT is also proposed to be involved in several of the hallmarks of cancer, the available information about its pro-tumoral role and mechanism of action is more limited. Here, we provide an overview of the involvement of both MIF and DDT in cancer, and we propose that blocking both cytokines is needed to obtain the maximum anti-tumor response.
Collapse
|
27
|
Jia D, Li B, Wang JK, Wang P, Li CY, Lu LX, Tian WY, Yu XH, Zhang JC, Zheng Y. Expression and Correlation of MIF and ERK1/2 in Liver Cirrhosis and Hepatocellular Carcinoma Induced by Hepatitis B. Pharmgenomics Pers Med 2023; 16:381-388. [PMID: 37124953 PMCID: PMC10145491 DOI: 10.2147/pgpm.s398976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/31/2023] [Indexed: 05/02/2023] Open
Abstract
Objective To detect expression and phosphorylation level of macrophage migration inhibitor (MIF) and extracellular-regulated kinases 1 and 2 (ERK1/2) in hepatitis B-induced liver cirrhosis (HBILC) and hepatocellular carcinoma (HCC) with a background of HBILC and analyze the correlation of MIF and ERK1/2 with HBILC and HCC. Methods Twenty cases of normal liver tissues were collected as a control group, and 48 specimens of HBILC tissues and 48 specimens of HCC tissues were collected as the experimental group, which were assigned as the HBILC group and HCC group, respectively. All tissue specimens were processed into tissue chips. The expressions of MIF, ERK1/2, and their phosphorylated proteins were detected via immunohistochemistry, and MIF and ERK1/2 nucleic acid expressions were detected by in situ hybridization. The results were statistically analyzed using the chi-square test. Results Proteins and nucleic acids of MIF and ERK1/2 presented low expression in the control group and high expression in the HBILC group and HCC group. MIF expression in the three groups was 25.0%, 75.0%, and 79.17%, respectively, while that of the nucleic acids was 25.0%, 70.83%, and 68.75%, respectively. Expression of ERK1/2 in the three groups was 40.0%, 60.42%, and 81.25%, respectively, and that of nucleic acids was 40.0%, 79.17%, and 77.08%. Expression of pERK1/2 was low in the control and HBILC group and high in the HCC group. Expression of pERK1/2 in the three groups was 20%, 45.83%, and 75%, respectively. Expression of pERK1/2 in the HCC group was significantly different from that in the HBILC and control group (P<0.05), but the difference between the HBILC group and control group was not statistically significant (P>0.05). Conclusion Occurrence and development of HBILC and HCC are not only related to the high expression of MIF but also closely related to the activation of the ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Dong Jia
- Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
| | - Bin Li
- Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
| | - Jun-Ke Wang
- Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
| | - Pan Wang
- Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
| | - Chu-Yi Li
- Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
| | - Li-Xia Lu
- Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
| | - Wen-Yan Tian
- Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
| | - Xiao-Hui Yu
- Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
- Correspondence: Xiao-Hui Yu; Jiu-Cong Zhang, Department of Gastroenterology, The 940 Hospital of Joint Logistic Support Force of People’s Liberation Army, No. 333 of Binhenan Road, Qilihe District, Lanzhou, 730050, People’s Republic of China, Tel +86 13919914665; +86 13919919690, Email ;
| | - Jiu-Cong Zhang
- Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
| | - Ying Zheng
- Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
| |
Collapse
|
28
|
Zan C, Yang B, Brandhofer M, El Bounkari O, Bernhagen J. D-dopachrome tautomerase in cardiovascular and inflammatory diseases-A new kid on the block or just another MIF? FASEB J 2022; 36:e22601. [PMID: 36269019 DOI: 10.1096/fj.202201213r] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 11/11/2022]
Abstract
Macrophage migration inhibitory factor (MIF) as well as its more recently described structural homolog D-dopachrome tautomerase (D-DT), now also termed MIF-2, are atypical cytokines and chemokines with key roles in host immunity. They also have an important pathogenic role in acute and chronic inflammatory conditions, cardiovascular diseases, lung diseases, adipose tissue inflammation, and cancer. Although our mechanistic understanding of MIF-2 is relatively limited compared to the extensive body of evidence available for MIF, emerging data suggests that MIF-2 is not only a functional phenocopy of MIF, but may have differential or even oppositional activities, depending on the disease and context. In this review, we summarize and discuss the similarities and differences between MIF and MIF-2, with a focus on their structures, receptors, signaling pathways, and their roles in diseases. While mainly covering the roles of the MIF homologs in cardiovascular, inflammatory, autoimmune, and metabolic diseases, we also discuss their involvement in cancer, sepsis, and chronic obstructive lung disease (COPD). A particular emphasis is laid upon potential mechanistic explanations for synergistic or cooperative activities of the MIF homologs in cancer, myocardial diseases, and COPD as opposed to emerging disparate or antagonistic activities in adipose tissue inflammation, metabolic diseases, and atherosclerosis. Lastly, we discuss potential future opportunities of jointly targeting MIF and MIF-2 in certain diseases, whereas precision targeting of only one homolog might be preferable in other conditions. Together, this article provides an update of the mechanisms and future therapeutic avenues of human MIF proteins with a focus on their emerging, surprisingly disparate activities, suggesting that MIF-2 displays a variety of activities that are distinct from those of MIF.
Collapse
Affiliation(s)
- Chunfang Zan
- Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU Klinikum, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Bishan Yang
- Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU Klinikum, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Markus Brandhofer
- Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU Klinikum, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Omar El Bounkari
- Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU Klinikum, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Jürgen Bernhagen
- Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU Klinikum, Ludwig-Maximilian-University (LMU), Munich, Germany.,Deutsches Zentrum für Herz-Kreislauferkrankungen (DZHK), Munich Heart Alliance, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
29
|
Sahu N, Grandi FC, Bhutani N. A single-cell mass cytometry platform to map the effects of preclinical drugs on cartilage homeostasis. JCI Insight 2022; 7:160702. [PMID: 36194485 PMCID: PMC9744259 DOI: 10.1172/jci.insight.160702] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 09/12/2022] [Indexed: 02/04/2023] Open
Abstract
No disease-modifying drug exists for osteoarthritis (OA). Despite success in animal models, candidate drugs continue to fail in clinical trials owing to the unmapped interpatient heterogeneity and disease complexity. We used a single-cell platform based on cytometry by time-of-flight (cyTOF) to precisely outline the effects of candidate drugs on human OA chondrocytes. OA chondrocytes harvested from patients undergoing total knee arthroplasty were treated with 2 drugs, an NF-κB pathway inhibitor, BMS-345541, and a chondroinductive small molecule, kartogenin, that showed preclinical success in animal models for OA. cyTOF conducted with 30 metal isotope-labeled antibodies parsed the effects of the drugs on inflammatory, senescent, and chondroprogenitor cell populations. The NF-κB pathway inhibition decreased the expression of p-NF-κB, HIF2A, and inducible NOS in multiple chondrocyte clusters and significantly depleted 4 p16ink4a-expressing senescent populations, including NOTCH1+STRO1+ chondroprogenitor cells. While kartogenin also affected select p16ink4a-expressing senescent clusters, there was a less discernible effect on chondroprogenitor cell populations. Overall, BMS-345541 elicited a uniform drug response in all patients, while only a few responded to kartogenin. These studies demonstrate that a single-cell cyTOF-based drug screening platform can provide insights into patient response assessment and patient stratification.
Collapse
|
30
|
Astaxanthin ameliorates serum level and spinal expression of macrophage migration inhibitory factor following spinal cord injury. Behav Pharmacol 2022; 33:505-512. [PMID: 36148838 DOI: 10.1097/fbp.0000000000000698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Astaxanthin (AST) is a lipid-soluble carotenoid with antioxidant and anti-inflammatory properties. Previous reports demonstrated the promising effects of AST on spinal cord injury (SCI)-induced inflammation and sensory-motor dysfunction. Macrophage migration inhibitory factor (MIF), as a cytokine, plays a critical role in the inflammatory phase of SCI. The aim of this study was to evaluate the effects of AST on post-SCI levels of MIF in serum and spinal cord. The possible correlation between MIF and mechanical pain threshold was also assessed. Adult male rats were subjected to a severe compression spinal injury and 30 min later were treated with AST (Intrathecal, 2 nmol) or vehicle. Neuropathic pain was assessed by von Frey filaments before the surgery, and then on days 7, 14, 21, and 28 post-SCI. Western blot and ELISA were used to measure the serum level and spinal expression of MIF following SCI in the same time points. AST treatment significantly attenuated the SCI-induced dysregulations in the serum levels and tissue expression of MIF. A negative correlation was observed between mechanical pain threshold and serum MIF level (r = -0.5463, P < 0.001), as well as mechanical pain threshold and spinal level of MIF (r = -0.9562; P < 0.001). AST ameliorates SCI-induced sensory dysfunction, probably through inhibiting MIF-regulated inflammatory pathways.
Collapse
|
31
|
Thiele M, Donnelly SC, Mitchell RA. OxMIF: a druggable isoform of macrophage migration inhibitory factor in cancer and inflammatory diseases. J Immunother Cancer 2022; 10:e005475. [PMID: 36180072 PMCID: PMC9528626 DOI: 10.1136/jitc-2022-005475] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2022] [Indexed: 11/04/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine with a pleiotropic spectrum of biological functions implicated in the pathogenesis of cancer and inflammatory diseases. MIF is constitutively present in several cell types and non-lymphoid tissues and is secreted after acute stress or inflammation. MIF triggers the release of proinflammatory cytokines, overrides the anti-inflammatory effects of glucocorticoids, and exerts chemokine function, resulting in increased migration and recruitment of leukocytes into inflamed tissue. Despite this, MIF is a challenging target for therapeutic intervention because of its ubiquitous nature and presence in the circulation and tissue of healthy individuals. Oxidized MIF (oxMIF) is an immunologically distinct disease-related structural isoform found in the plasma and tissues of patients with inflammatory diseases and in solid tumor tissues. MIF converts to oxMIF in an oxidizing, inflammatory environment. This review discusses the biology and activity of MIF and the potential for autoimmune disease and cancer modification by targeting oxMIF. Anti-oxMIF antibodies reduce cancer cell invasion/migration, angiogenesis, proinflammatory cytokine production, and ERK and AKT activation. Anti-oxMIF antibodies also elicit apoptosis and alter immune cell function and/or migration. When co-administered with a glucocorticoid, anti-oxMIF antibodies produced a synergistic response in inflammatory models. Anti-oxMIF antibodies therefore counterregulate biological activities attributed to MIF. oxMIF expression has been observed in inflammatory diseases (eg, sepsis, psoriasis, asthma, inflammatory bowel disease, and systemic lupus erythematosus) and oxMIF has been detected in ovarian, colorectal, lung, and pancreatic cancers. In contrast to MIF, oxMIF is specifically detected in plasma and/or tissues of diseased patients, but not in healthy individuals. Therefore, as a druggable isoform of MIF, oxMIF represents a potential new therapeutic target in inflammatory diseases and cancer. Fully human, monoclonal anti-oxMIF antibodies have been shown to selectively bind oxMIF in preclinical and phase I studies; however, additional clinical assessments are necessary to validate their use as either a monotherapy or in combination with standard-of-care regimens (ie, immunomodulatory agents/checkpoint inhibitors, anti-angiogenic drugs, chemotherapeutics, and glucocorticoids).
Collapse
Affiliation(s)
- Michael Thiele
- Biology Research, OncoOne Research & Development GmbH, Vienna, Austria
| | - Seamas C Donnelly
- Department of Medicine, Tallaght University Hospital & Trinity College Dublin, Dublin, Ireland
| | - Robert A Mitchell
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky, USA
- Department of Surgery, J.G. Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
- Division of Immunotherapy, Department of Surgery, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
32
|
Wang D, Min S, Lin X, Jiang G. Association Among MIF, IFIH1, and IL6 Gene Polymorphisms and Non-Segmental Vitiligo in a Chinese Han Population. Clin Cosmet Investig Dermatol 2022; 15:1597-1609. [PMID: 35983127 PMCID: PMC9380431 DOI: 10.2147/ccid.s369418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/13/2022] [Indexed: 11/23/2022]
Abstract
Objective The aim of the present study was to investigate the association of single-nucleotide polymorphisms (SNPs) in the macrophage migration inhibiting factor (MIF), interferon-induced Helicase C domain 1 (IFIH1), interleukin-6 (IL6) genes, circulating levels with non-segmental vitiligo (NSV) susceptibility in the Chinese population, and to analyze the relationships between gene polymorphisms and clinical characteristics of vitiligo. Methods In this study, genotyping was conducted in 155 patients with NSV and 117 unaffected controls using polymerase chain reaction and snapshot technique. Serum concentrations were determined by ELISA kit. Results There were strong associations between IFIH1 H843R and IL6-572G/C polymorphisms and NSV susceptibility (p = 0.013; p = 0.009). In contrast to previous studies, we found no significant difference in the MIF-173G/C polymorphism between the two groups. In addition, the frequency of allelic distribution for MIF-173G/C in patients with active NSV was significantly higher than stable NSV (p = 0.011), and IFIH1 H843R with early-onset (≤ 20), active or family history of NSV was significantly higher than late-onset (> 20), stable or no family history of NSV (p = 0.033; p = 0.045; p = 0.039). Serum concentrations of MIF were higher in patients with active NSV, serum IFIH1 and IL6 concentrations were related to the presence of polymorphisms in patients with NSV (p = 0.009; p = 0.011). Conclusion Our results suggested that IFIH1 H843R and IL6-572G/C gene polymorphisms and expression levels are obviously correlated with the onset of NSV. MIF-173G/C allele and serum concentrations may be associated with active NSV, and IFIH1 H843R allele may be associated with youth, active or family history of NSV.
Collapse
Affiliation(s)
- Danfeng Wang
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,First Clinical Medical College, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Shuhui Min
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,First Clinical Medical College, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Xiao Lin
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,First Clinical Medical College, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Guan Jiang
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,First Clinical Medical College, Xuzhou Medical University, Xuzhou, People's Republic of China
| |
Collapse
|