1
|
He F, Xu J, Zeng F, Wang B, Yang Y, Xu J, Sun X, Ren T, Tang X. Integrative analysis of Ewing's sarcoma reveals that the MIF-CD74 axis is a target for immunotherapy. Cell Commun Signal 2025; 23:23. [PMID: 39800691 PMCID: PMC11727170 DOI: 10.1186/s12964-024-02020-y] [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: 10/21/2024] [Accepted: 12/28/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Ewing's sarcoma (EwS), a common pediatric bone cancer, is associated with poor survival due to a lack of therapeutic targets for immunotherapy or targeted therapy. Therefore, more effective treatment options are urgently needed. METHODS Since novel immunotherapies may address this need, we performed an integrative analysis involving single-cell RNA sequencing, cell function experiments, and humanized models to dissect the immunoregulatory interactions in EwS and identify strategies for optimizing immunotherapeutic efficacy. RESULTS EwS is infiltrated by immunosuppressive myeloid populations, T and B lymphocytes, and natural killer cells. We found that SLC40A1 and C1QA macrophages were associated with a poor prognosis, whereas CD8+ T-cell infiltration was associated with a good prognosis. A comparative analysis of paired samples revealed that in tumors with a good chemotherapeutic response, macrophages presented increased antigen presentation and reduced release of protumor cytokines, whereas CD8+ T cells presented increased cytotoxicity and reduced exhaustion. An interaction analysis revealed a vast immunoregulatory network and identified MIF-CD74 as a crucial immunoregulatory target that can simultaneously promote M2 polarization of macrophages and inhibit CD8+ T-cell infiltration. Importantly, MIF blockade effectively reshaped the tumor immune microenvironment, turning cold tumors hot and inhibiting tumor growth. CONCLUSIONS Our integrative analysis revealed that the MIF/CD74 axis is a promising target for the treatment of Ewing sarcoma and provides a rationale for this novel immunotherapy.
Collapse
MESH Headings
- Sarcoma, Ewing/immunology
- Sarcoma, Ewing/therapy
- Sarcoma, Ewing/pathology
- Sarcoma, Ewing/metabolism
- Sarcoma, Ewing/genetics
- Humans
- Macrophage Migration-Inhibitory Factors/metabolism
- Immunotherapy
- Histocompatibility Antigens Class II/metabolism
- Histocompatibility Antigens Class II/immunology
- Antigens, Differentiation, B-Lymphocyte/metabolism
- Antigens, Differentiation, B-Lymphocyte/immunology
- Intramolecular Oxidoreductases/metabolism
- Intramolecular Oxidoreductases/genetics
- Animals
- Macrophages/immunology
- Macrophages/metabolism
- Cell Line, Tumor
- CD8-Positive T-Lymphocytes/immunology
- Bone Neoplasms/immunology
- Bone Neoplasms/pathology
- Bone Neoplasms/therapy
- Bone Neoplasms/metabolism
- Mice
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Fangzhou He
- Department of Musculoskeletal Tumor, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Jiuhui Xu
- Department of Musculoskeletal Tumor, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Fanwei Zeng
- Department of Musculoskeletal Tumor, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Boyang Wang
- Department of Musculoskeletal Tumor, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Yi Yang
- Department of Musculoskeletal Tumor, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, China
| | - Jie Xu
- Department of Musculoskeletal Tumor, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, China
| | - Xin Sun
- Department of Musculoskeletal Tumor, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, China
| | - Tingting Ren
- Department of Musculoskeletal Tumor, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, China.
- Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China.
| | - Xiaodong Tang
- Department of Musculoskeletal Tumor, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, China.
- Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China.
| |
Collapse
|
2
|
Al Shboul S, Singh A, Kobetic R, Goodlett DR, Brennan PM, Hupp T, Dapic I. Mass Spectrometry Advances in Analysis of Glioblastoma. MASS SPECTROMETRY REVIEWS 2024. [PMID: 39529217 DOI: 10.1002/mas.21912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/06/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Some cancers such as glioblastoma (GBM), show minimal response to medical interventions, often only capable of mitigating tumor growth or alleviating symptoms. High metabolic activity in the tumor microenvironment marked by immune responses and hypoxia, is a crucial factor driving tumor progression. The many developments in mass spectrometry (MS) over the last decades have provided a pivotal tool for studying proteins, along with their posttranslational modifications. It is known that the proteomic landscape of GBM comprises a wide range of proteins involved in cell proliferation, survival, migration, and immune evasion. Combination of MS imaging and microscopy has potential to reveal the spatial and molecular characteristics of pathological tissue sections. Moreover, integration of MS in the surgical process in form of techniques such as DESI-MS or rapid evaporative ionization MS has been shown as an effective tool for rapid measurement of metabolite profiles, providing detailed information within seconds. In immunotherapy-related research, MS plays an indispensable role in detection and targeting of cancer antigens which serve as a base for antigen-specific therapies. In this review, we aim to provide detailed information on molecular profile in GBM and to discuss recent MS advances and their clinical benefits for targeting this aggressive disease.
Collapse
Affiliation(s)
- Sofian Al Shboul
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Ashita Singh
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, UK
| | | | - David R Goodlett
- University of Victoria-Genome BC Proteomics Centre, Victoria, British Columbia, Canada
| | - Paul M Brennan
- Translational Neurosurgery, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Ted Hupp
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, UK
| | | |
Collapse
|
3
|
Ryan N, Lamenza F, Shrestha S, Upadhaya P, Springer A, Jordanides P, Pracha H, Roth P, Kumar R, Wang Y, Vilgelm AE, Satoskar A, Oghumu S. Host derived macrophage migration inhibitory factor expression attenuates anti-tumoral immune cell accumulation and promotes immunosuppression in the tumor microenvironment of head and neck squamous cell carcinoma. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167345. [PMID: 38992847 DOI: 10.1016/j.bbadis.2024.167345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/24/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a significant public health concern worldwide. Immunomodulatory targets in the HNSCC tumor microenvironment are crucial to enhance the efficacy of HNSCC immunotherapy. Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine that has been linked to poor prognosis in many cancers, but the mechanistic role of MIF in HNSCC remains unclear. Using a murine orthotopic oral cancer model in Mif+/+ or Mif-/- mice, we determined the function of host derived MIF in HNSCC tumor development, metastasis as well as localized and systemic tumor immune responses. We observed that Mif-/- mice have decreased tumor growth and tumor burden compared to their wild-type counterparts. Flow cytometric analysis of immune populations within the primary tumor site revealed increased Th1 and cytotoxic T cell recruitment to the HNSCC tumor microenvironment. Within the tumors of Mif-/- mice, MIF deletion also enhanced the effector function of anti-tumoral effector CD8+ T cells as well as Th1 cells and decreased the accumulation of granulocytic myeloid derived suppressor cells (g-MDSCs) in the tumor microenvironment. Furthermore, MDSCs isolated from tumor bearing mice chemotactically respond to MIF in a dose dependent manner. Taken together, our results demonstrate a chemotactic and immunomodulatory role for host derived MIF in promoting HNSCC and suggest that MIF targeted immunomodulation is a promising approach for HNSCC treatment.
Collapse
Affiliation(s)
- Nathan Ryan
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; College of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Felipe Lamenza
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Suvekshya Shrestha
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Puja Upadhaya
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Anna Springer
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Pete Jordanides
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Hasan Pracha
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Peyton Roth
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Rathan Kumar
- Department of Hematology, The Ohio State University Wexner Medial Center, Columbus, OH 43210, USA
| | - Yinchong Wang
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Anna E Vilgelm
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Abhay Satoskar
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Steve Oghumu
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
4
|
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
|
5
|
Fey RM, Nichols RA, Tran TT, Vandenbark AA, Kulkarni RP. MIF and CD74 as Emerging Biomarkers for Immune Checkpoint Blockade Therapy. Cancers (Basel) 2024; 16:1773. [PMID: 38730725 PMCID: PMC11082995 DOI: 10.3390/cancers16091773] [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: 03/14/2024] [Revised: 04/18/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Immune checkpoint blockade (ICB) therapy is used to treat a wide range of cancers; however, some patients are at risk of developing treatment resistance and/or immune-related adverse events (irAEs). Thus, there is a great need for the identification of reliable predictive biomarkers for response and toxicity. The cytokine MIF (macrophage migration inhibitory factor) and its cognate receptor CD74 are intimately connected with cancer progression and have previously been proposed as prognostic biomarkers for patient outcome in various cancers, including solid tumors such as malignant melanoma. Here, we assess their potential as predictive biomarkers for response to ICB therapy and irAE development. We provide a brief overview of their function and roles in the context of cancer and autoimmune disease. We also review the evidence showing that MIF and CD74 may be of use as predictive biomarkers of patient response to ICB therapy and irAE development. We also highlight that careful consideration is required when assessing the potential of serum MIF levels as a biomarker due to its reported circadian expression in human plasma. Finally, we suggest future directions for the establishment of MIF and CD74 as predictive biomarkers for ICB therapy and irAE development to guide further research in this field.
Collapse
Affiliation(s)
- Rosalyn M. Fey
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA (R.A.N.)
| | - Rebecca A. Nichols
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA (R.A.N.)
| | - Thuy T. Tran
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Arthur A. Vandenbark
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, Portland, OR 97239, USA
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rajan P. Kulkarni
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA (R.A.N.)
- Cancer Early Detection Advanced Research Center (CEDAR), Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
- Operative Care Division, U.S. Department of Veterans Affairs Portland Health Care System, Portland, OR 97239, USA
| |
Collapse
|
6
|
Valdez CN, Sánchez-Zuno GA, Bucala R, Tran TT. Macrophage Migration Inhibitory Factor (MIF) and D-Dopachrome Tautomerase (DDT): Pathways to Tumorigenesis and Therapeutic Opportunities. Int J Mol Sci 2024; 25:4849. [PMID: 38732068 PMCID: PMC11084905 DOI: 10.3390/ijms25094849] [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: 03/28/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Discovered as inflammatory cytokines, MIF and DDT exhibit widespread expression and have emerged as critical mediators in the response to infection, inflammation, and more recently, in cancer. In this comprehensive review, we provide details on their structures, binding partners, regulatory mechanisms, and roles in cancer. We also elaborate on their significant impact in driving tumorigenesis across various cancer types, supported by extensive in vitro, in vivo, bioinformatic, and clinical studies. To date, only a limited number of clinical trials have explored MIF as a therapeutic target in cancer patients, and DDT has not been evaluated. The ongoing pursuit of optimal strategies for targeting MIF and DDT highlights their potential as promising antitumor candidates. Dual inhibition of MIF and DDT may allow for the most effective suppression of canonical and non-canonical signaling pathways, warranting further investigations and clinical exploration.
Collapse
Affiliation(s)
- Caroline Naomi Valdez
- School of Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA; (C.N.V.); (R.B.)
| | - Gabriela Athziri Sánchez-Zuno
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA;
| | - Richard Bucala
- School of Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA; (C.N.V.); (R.B.)
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA;
- Yale Cancer Center, Yale University, 333 Cedar St., New Haven, CT 06510, USA
| | - Thuy T. Tran
- School of Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA; (C.N.V.); (R.B.)
- Yale Cancer Center, Yale University, 333 Cedar St., New Haven, CT 06510, USA
- Section of Medical Oncology, Department of Internal Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA
| |
Collapse
|
7
|
Fang T, Liu L, Song D, Huang D. The role of MIF in periodontitis: A potential pathogenic driver, biomarker, and therapeutic target. Oral Dis 2024; 30:921-937. [PMID: 36883414 DOI: 10.1111/odi.14558] [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: 01/03/2023] [Revised: 02/08/2023] [Accepted: 03/01/2023] [Indexed: 03/09/2023]
Abstract
OBJECTIVE Periodontitis is an inflammatory disease that involves an imbalance in the oral microbiota, activation of inflammatory and immune responses, and alveolar bone destruction. Macrophage migration inhibitory factor (MIF) is a versatile cytokine involved in several pathological reactions, including inflammatory processes and bone destruction, both of which are characteristics of periodontitis. While the roles of MIF in cancer and other immune diseases have been extensively characterized, its role in periodontitis remains inconclusive. RESULTS In this review, we describe a comprehensive analysis of the potential roles of MIF in periodontitis from the perspective of immune response and bone regulation at the cellular and molecular levels. Moreover, we discuss its potential reliability as a novel diagnostic and therapeutic target for periodontitis. CONCLUSION This review can aid dental researchers and clinicians in understanding the current state of MIF-related pathogenesis, diagnosis, and treatment of periodontitis.
Collapse
Affiliation(s)
- Tongfeng Fang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liu Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dongzhe Song
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dingming Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
8
|
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
|
9
|
Jarmula J, Lee J, Lauko A, Rajappa P, Grabowski MM, Dhawan A, Chen P, Bucala R, Vogelbaum MA, Lathia JD. Macrophage migration inhibitory factor as a therapeutic target in neuro-oncology: A review. Neurooncol Adv 2024; 6:vdae142. [PMID: 39233830 PMCID: PMC11372298 DOI: 10.1093/noajnl/vdae142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024] Open
Abstract
Primary central nervous system (CNS) tumors affect tens of thousands of patients each year, and there is a significant need for new treatments. Macrophage migration inhibitory factor (MIF) is a cytokine implicated in multiple tumorigenic processes such as cell proliferation, vascularization, and immune evasion and is therefore a promising therapeutic target in primary CNS tumors. There are several MIF-directed treatments available, including small-molecule inhibitors, peptide drugs, and monoclonal antibodies. However, only a small number of these drugs have been tested in preclinical models of primary CNS tumors, and even fewer have been studied in patients. Moreover, the brain has unique therapeutic requirements that further make effective targeting challenging. In this review, we summarize the latest functions of MIF in primary CNS tumor initiation and progression. We also discuss advances in MIF therapeutic development and ongoing preclinical studies and clinical trials. Finally, we discuss potential future MIF therapies and the strategies required for successful clinical translation.
Collapse
Affiliation(s)
- Jakub Jarmula
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
| | - Juyeun Lee
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Adam Lauko
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
| | - Prajwal Rajappa
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Matthew M Grabowski
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Andrew Dhawan
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Peiwen Chen
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Richard Bucala
- Section of Rheumatology, Allergy, and Immunology, Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Michael A Vogelbaum
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Justin D Lathia
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
10
|
Zhang WX, Huang J, Tian XY, Liu YH, Jia MQ, Wang W, Jin CY, Song J, Zhang SY. A review of progress in o-aminobenzamide-based HDAC inhibitors with dual targeting capabilities for cancer therapy. Eur J Med Chem 2023; 259:115673. [PMID: 37487305 DOI: 10.1016/j.ejmech.2023.115673] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/10/2023] [Accepted: 07/19/2023] [Indexed: 07/26/2023]
Abstract
Histone deacetylases, as a new class of anticancer targets, could maintain homeostasis by catalyzing histone deacetylation and play important roles in regulating the expression of target genes. Due to the fact that simultaneous intervention with dual tumor related targets could improve treatment effects, researches on innovative design of dual-target drugs are underway. HDAC is known as a "sensitizer" for the synergistic effects with other anticancer-target drugs because of its flexible structure design. The synergistic effects of HDAC inhibitor and other target inhibitors usually show enhanced inhibitory effects on tumor cells, and also provide new strategies to overcome multidrug resistance. Many research groups have reported that simultaneously inhibiting HDAC and other targets, such as tubulin, EGFR, could enhance the therapeutic effects. The o-aminobenzamide group is often used as a ZBG group in the design of HDAC inhibitors with potent antitumor effects. Given the prolonged inhibitory effects and reduced toxic side effects of HDAC inhibitors using o-aminobenzamide as the ZBG group, the o-aminobenzamide group is expected to become a more promising alternative to hydroxamic acid. In fact, o-aminobenzamide-based dual inhibitors of HDAC with different chemical structures have been extensively prepared and reported with synergistic and enhanced anti-tumor effects. In this work, we first time reviewed the rational design, molecular docking, inhibitory activities and potential application of o-aminobenzamide-based HDAC inhibitors with dual targeting capabilities in cancer therapy, which might provide a reference for developing new and more effective anticancer drugs.
Collapse
Affiliation(s)
- Wei-Xin Zhang
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Jiao Huang
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Xin-Yi Tian
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yun-He Liu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Mei-Qi Jia
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wang Wang
- Luoyang Key Laboratory of Organic Functional Molecules, College of Food and Drug, Luoyang Normal University, Luoyang, 471934, China
| | - Cheng-Yun Jin
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Jian Song
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Sai-Yang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
11
|
Hasib RA, Ali MC, Rahman MH, Ahmed S, Sultana S, Summa SZ, Shimu MSS, Afrin Z, Jamal MAHM. Integrated gene expression profiling and functional enrichment analyses to discover biomarkers and pathways associated with Guillain-Barré syndrome and autism spectrum disorder to identify new therapeutic targets. J Biomol Struct Dyn 2023; 42:11299-11321. [PMID: 37776011 DOI: 10.1080/07391102.2023.2262586] [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: 03/15/2023] [Accepted: 09/17/2023] [Indexed: 10/01/2023]
Abstract
Guillain-Barré syndrome (GBS) is one of the most prominent and acute immune-mediated peripheral neuropathy, while autism spectrum disorders (ASD) are a group of heterogeneous neurodevelopmental disorders. The complete mechanism regarding the neuropathophysiology of these disorders is still ambiguous. Even after recent breakthroughs in molecular biology, the link between GBS and ASD remains a mystery. Therefore, we have implemented well-established bioinformatic techniques to identify potential biomarkers and drug candidates for GBS and ASD. 17 common differentially expressed genes (DEGs) were identified for these two disorders, which later guided the rest of the research. Common genes identified the protein-protein interaction (PPI) network and pathways associated with both disorders. Based on the PPI network, the constructed hub gene and module analysis network determined two common DEGs, namely CXCL9 and CXCL10, which are vital in predicting the top drug candidates. Furthermore, coregulatory networks of TF-gene and TF-miRNA were built to detect the regulatory biomolecules. Among drug candidates, imatinib had the highest docking and MM-GBSA score with the well-known chemokine receptor CXCR3 and remained stable during the 100 ns molecular dynamics simulation validated by the principal component analysis and the dynamic cross-correlation map. This study predicted the gene-based disease network for GBS and ASD and suggested prospective drug candidates. However, more in-depth research is required for clinical validation.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rizone Al Hasib
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia, Bangladesh
- Laboratory of Medical and Environmental Biotechnology Islamic University, Kushtia, Bangladesh
| | - Md Chayan Ali
- Department of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Md Habibur Rahman
- Department of Computer Science and Engineering, Islamic University, Kushtia, Bangladesh
- Center for Advanced Bioinformatics and Artificial Intelligent Research, Islamic University, Kushtia, Bangladesh
| | - Sabbir Ahmed
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia, Bangladesh
| | - Shaharin Sultana
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia, Bangladesh
- Laboratory of Medical and Environmental Biotechnology Islamic University, Kushtia, Bangladesh
| | - Sadia Zannat Summa
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia, Bangladesh
- Laboratory of Medical and Environmental Biotechnology Islamic University, Kushtia, Bangladesh
| | | | - Zinia Afrin
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia, Bangladesh
| | - Mohammad Abu Hena Mostofa Jamal
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia, Bangladesh
- Laboratory of Medical and Environmental Biotechnology Islamic University, Kushtia, Bangladesh
| |
Collapse
|
12
|
Sooreshjani M, Tripathi S, Dussold C, Najem H, de Groot J, Lukas RV, Heimberger AB. The Use of Targeted Cytokines as Cancer Therapeutics in Glioblastoma. Cancers (Basel) 2023; 15:3739. [PMID: 37509400 PMCID: PMC10378451 DOI: 10.3390/cancers15143739] [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: 06/02/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Cytokines play an important role in regulating the immune response. Although there is great interest in exploiting cytokines for cancer immunotherapy, their clinical potential is limited by their pleiotropic properties and instability. A variety of cancer cell-intrinsic and extrinsic characteristics pose a barrier to effective treatments including cytokines. Recent studies using gene and cell therapy offer new opportunities for targeting cytokines or their receptors, demonstrating that they are actionable targets. Current efforts such as virotherapy, systemic cytokine therapy, and cellular and gene therapy have provided novel strategies that incorporate cytokines as potential therapeutic strategies for glioblastoma. Ongoing research on characterizing the tumor microenvironment will be informative for prioritization and combinatorial strategies of cytokines for future clinical trials. Unique therapeutic opportunities exist at the convergence of cytokines that play a dual role in tumorigenesis and immune modulation. Here, we discuss the underlying strategies in pre- and clinical trials aiming to enhance treatment outcomes in glioblastoma patients.
Collapse
Affiliation(s)
- Moloud Sooreshjani
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Shashwat Tripathi
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Corey Dussold
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Hinda Najem
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - John de Groot
- Department of Neurosurgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Rimas V. Lukas
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Amy B. Heimberger
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neurosurgery, Northwestern University, Chicago, IL60611, USA
- Simpson Querrey Biomedical Research Center, 303 E. Superior Street, 6-516, Chicago, IL 60611, USA
| |
Collapse
|
13
|
Ebert S, Zang L, Ismail N, Otabil M, Fröhlich A, Egea V, Ács S, Hoeberg M, Berres ML, Weber C, Moreira JMA, Ries C, Bernhagen J, El Bounkari O. Tissue Inhibitor of Metalloproteinases-1 Interacts with CD74 to Promote AKT Signaling, Monocyte Recruitment Responses, and Vascular Smooth Muscle Cell Proliferation. Cells 2023; 12:1899. [PMID: 37508563 PMCID: PMC10378328 DOI: 10.3390/cells12141899] [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: 06/13/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Tissue inhibitor of metalloproteinases-1 (TIMP-1), an important regulator of matrix metalloproteinases (MMPs), has recently been shown to interact with CD74, a receptor for macrophage migration inhibitory factor (MIF). However, the biological effects mediated by TIMP-1 through CD74 remain largely unexplored. Using sequence alignment and in silico protein-protein docking analysis, we demonstrated that TIMP-1 shares residues with both MIF and MIF-2, crucial for CD74 binding, but not for CXCR4. Subcellular colocalization, immunoprecipitation, and internalization experiments supported these findings, demonstrating that TIMP-1 interacts with surface-expressed CD74, resulting in its internalization in a dose-dependent manner, as well as with a soluble CD74 ectodomain fragment (sCD74). This prompted us to study the effects of the TIMP-1-CD74 axis on monocytes and vascular smooth muscle cells (VSCMs) to assess its impact on vascular inflammation. A phospho-kinase array revealed the activation of serine/threonine kinases by TIMP-1 in THP-1 pre-monocytes, in particular AKT. Similarly, TIMP-1 dose-dependently triggered the phosphorylation of AKT and ERK1/2 in primary human monocytes. Importantly, Transwell migration, 3D-based Chemotaxis, and flow adhesion assays demonstrated that TIMP-1 engagement of CD74 strongly promotes the recruitment response of primary human monocytes, while live cell imaging studies revealed a profound activating effect on VSMC proliferation. Finally, re-analysis of scRNA-seq data highlighted the expression patterns of TIMP-1 and CD74 in human atherosclerotic lesions, thus, together with our experimental data, indicating a role for the TIMP-1-CD74 axis in vascular inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Simon Ebert
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 81377 Munich, Germany
| | - Lan Zang
- Institute for Cardiovascular Prevention (IPEK), Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 80336 Munich, Germany
| | - Noor Ismail
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 81377 Munich, Germany
| | - Michael Otabil
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 81377 Munich, Germany
| | - Adrian Fröhlich
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 81377 Munich, Germany
| | - Virginia Egea
- Institute for Cardiovascular Prevention (IPEK), Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 80336 Munich, Germany
| | - Susann Ács
- Institute for Cardiovascular Prevention (IPEK), Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 80336 Munich, Germany
| | - Mikkel Hoeberg
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Marie-Luise Berres
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 80336 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Munich Heart Alliance, 80802 Munich, Germany
| | - José M A Moreira
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christian Ries
- Institute for Cardiovascular Prevention (IPEK), Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 80336 Munich, Germany
| | - Jürgen Bernhagen
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Munich Heart Alliance, 80802 Munich, Germany
| | - Omar El Bounkari
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 81377 Munich, Germany
| |
Collapse
|
14
|
Montemurro N, Pahwa B, Tayal A, Shukla A, De Jesus Encarnacion M, Ramirez I, Nurmukhametov R, Chavda V, De Carlo A. Macrophages in Recurrent Glioblastoma as a Prognostic Factor in the Synergistic System of the Tumor Microenvironment. Neurol Int 2023; 15:595-608. [PMID: 37218976 DOI: 10.3390/neurolint15020037] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/05/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Glioblastoma (GBM) is a common and highly malignant primary tumor of the central nervous system in adults. Ever more recent papers are focusing on understanding the role of the tumor microenvironment (TME) in affecting tumorigenesis and the subsequent prognosis. We assessed the impact of macrophages in the TME on the prognosis in patients with recurrent GBM. A PubMed, MEDLINE and Scopus review was conducted to identify all studies dealing with macrophages in the GBM microenvironment from January 2016 to December 2022. Glioma-associated macrophages (GAMs) act critically in enhancing tumor progression and can alter drug resistance, promoting resistance to radiotherapy and establishing an immunosuppressive environment. M1 macrophages are characterized by increased secretion of proinflammatory cytokines, such as IL-1ß, tumor necrosis factor (TNF), IL-27, matrix metalloproteinase (MMPs), CCL2, and VEGF (vascular endothelial growth factor), IGF1, that can lead to the destruction of the tissue. In contrast, M2 is supposed to participate in immunosuppression and tumor progression, which is formed after being exposed to the macrophage M-CSF, IL-10, IL-35 and the transforming growth factor-ß (TGF-β). Because there is currently no standard of care in recurrent GBM, novel identified targeted therapies based on the complex signaling and interactions between the glioma stem cells (GSCs) and the TME, especially resident microglia and bone-marrow-derived macrophages, may be helpful in improving the overall survival of these patients in the near future.
Collapse
Affiliation(s)
- Nicola Montemurro
- Department of Neurosurgery, Azienda Ospedaliero Universitaria Pisana (AOUP), University of Pisa, 56100 Pisa, Italy
| | - Bhavya Pahwa
- University College of Medical Sciences and GTB Hospital, New Delhi 110095, India
| | - Anish Tayal
- University College of Medical Sciences and GTB Hospital, New Delhi 110095, India
| | - Anushruti Shukla
- University College of Medical Sciences and GTB Hospital, New Delhi 110095, India
| | | | - Issael Ramirez
- Royal Melbourne Hospital, Melbourne, VIC 3000, Australia
| | - Renat Nurmukhametov
- Department of Spinal Surgery, Central Clinical Hospital of the Russian Academy of Sciences, 121359 Moscow, Russia
| | - Vishal Chavda
- Department of Pathology, Stanford of School of Medicine, Stanford University Medical Centre, Palo Alto, CA 94305, USA
| | - Antonella De Carlo
- Department of Neurosurgery, Azienda Ospedaliero Universitaria Pisana (AOUP), University of Pisa, 56100 Pisa, Italy
| |
Collapse
|
15
|
Metabolic reprograming of MDSCs within tumor microenvironment and targeting for cancer immunotherapy. Acta Pharmacol Sin 2022; 43:1337-1348. [PMID: 34561553 PMCID: PMC9160034 DOI: 10.1038/s41401-021-00776-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023] Open
Abstract
A number of emerging studies in field of immune metabolism have indicated that cellular metabolic reprograming serves as a major administrator in maintaining the viability and functions of both tumor cells and immune cells. As one of the most important immunosuppressive cells in tumor stroma, myeloid-derived suppressor cells (MDSCs) dynamically orchestrate their metabolic pathways in response to the complicated tumor microenvironment (TME), a process that consequently limits the therapeutic effectiveness of anti-cancer treatment modalities. In this context, the metabolic vulnerabilities of MDSCs could be exploited as a novel immune metabolic checkpoint upon which to intervene for promoting the efficacy of immunotherapy. Here, we have discussed about recent studies highlighting the important roles of the metabolic reprograming and the core molecular pathways involved in tumor-infiltrating MDSCs. In addition, we have also summarized the state-of-the-art strategies that are currently being employed to target MDSC metabolism and improve the efficacy of antineoplastic immunotherapy.
Collapse
|
16
|
Qu B, Han X, Zhao L, Zhang F, Gao Q. Relationship of HIF‑1α expression with apoptosis and cell cycle in bone marrow mesenchymal stem cells from patients with myelodysplastic syndrome. Mol Med Rep 2022; 26:239. [PMID: 35642674 PMCID: PMC9185697 DOI: 10.3892/mmr.2022.12755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 02/21/2022] [Indexed: 11/09/2022] Open
Abstract
Myelodysplastic syndrome (MDS) is a group of abnormal clonal disorders with ineffective hematopoiesis, which are incurable with conventional therapy. Of note, MDS features an abnormal bone marrow microenvironment, which is related to its incidence. The hypoxia-inducible factor-1α (HIF-1α) transcriptional signature is generally activated in bone marrow stem/progenitor cells of patients with MDS. To analyze the expression of HIF-1α in bone marrow mesenchymal stem cells (BM-MSCs) and the apoptosis and cell cycle features associated with the disease, BM-MSCs were obtained from 40 patients with a definitive diagnosis of MDS and 20 subjects with hemocytopenia but a negative diagnosis of MDS as a control group. Reverse transcription-quantitative PCR and western blot analyses were used to measure HIF-1α expression in cells from the two groups and apoptosis and cell cycle were also analyzed and compared between the groups using flow cytometry assays. BM-MSCs from both the control group and the MDS group exhibited a fibroblast-like morphology, had similar growth cycles and were difficult to passage stably. It was observed that BM-MSCs from the MDS group had significantly higher HIF-1α expression levels than the control group (P<0.05). Furthermore, the BM-MSCs from the MDS group had a higher proportion of cells in early apoptosis (5.22±1.34 vs. 2.04±0.08%; P<0.0001) and late apoptosis (3.38±0.43 vs. 1.23±0.11%; P<0.01) and exhibited cell cycle arrest. This may be a noteworthy aspect of the pathogenesis of MDS and may be related to high HIF-1α expression under a hypoxic state in the bone marrow microenvironment. Furthermore, the expression of HIF-1α in bone marrow tissue sections from patients with MDS in the International Prognostic Scoring System (IPSS) lower-risk group was higher than that from patients with MDS in the IPSS high-risk group. These results revealed the role of HIF-1α as a central pathobiology mediator of MDS and an effective therapeutic target for a broad spectrum of patients with MDS, particularly for patients in the lower-risk group.
Collapse
Affiliation(s)
- Beibei Qu
- Department of Hematology, Jiading District Central Hospital, Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201800, P.R. China
| | - Xiuhua Han
- Department of Hematology, Jiading District Central Hospital, Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201800, P.R. China
| | - Lan Zhao
- Department of Hematology, Jiading District Central Hospital, Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201800, P.R. China
| | - Feifei Zhang
- Department of Hematology, Jiading District Central Hospital, Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201800, P.R. China
| | - Qingmei Gao
- Department of Hematology, Jiading District Central Hospital, Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201800, P.R. China
| |
Collapse
|
17
|
Bielecka-Wajdman AM, Ludyga T, Smyk D, Smyk W, Mularska M, Świderek P, Majewski W, Mullins CS, Linnebacher M, Obuchowicz E. Glucose Influences the Response of Glioblastoma Cells to Temozolomide and Dexamethasone. Cancer Control 2022; 29:10732748221075468. [PMID: 35225010 PMCID: PMC8891890 DOI: 10.1177/10732748221075468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Objective Current research indicates that weakness of glucose metabolism plays an important role in silencing of invasiveness and growth of hypoxic tumors such as GBM. Moreover, there are indications that DXM, frequently used in treatment, may support GBM energy metabolism and provoke its recurrence. Methods We carried out in vitro experiments on the commercial T98G cell line and two primary GBM lines (HROG02, HROG17) treated with TMZ and/or DXM in physiological oxygen conditions for GBM (2.5% oxygen) and for comparison, in standard laboratory conditions (20% oxygen). The influence of different glucose levels on selected malignancy features of GBM cells-cellular viability and division, dynamic of cell culture changes, colony formation and concentration of InsR have been elevated. Results Under 2.5% oxygen and high glucose concentration, an attenuated cytotoxic effect of TMZ and intensification of malignancy features in all glioblastoma cell lines exposed to DXM was seen. Furthermore, preliminary retrospective analysis to assess the correlation between serum glucose levels and Ki-67 expression in surgical specimens derived from patients with GBM (IV) treated with radio-chemotherapy and prophylactic DXM therapy was performed. Conclusion The data suggest a link between the in vitro study results and clinical data. High glucose can influence on GBM progression through the promotion of the following parameters: cell viability, dispersal, InsR expression and cell proliferation (Ki-67). However, this problem needs more studies and explain the mechanism of action studied drugs.
Collapse
Affiliation(s)
- Anna M Bielecka-Wajdman
- Department of Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
- Anna Bielecka-Wajdman, Department of Pharmacology, Medical University of Silesia, Medyków 18, Katowice 40-055, Poland.
| | - Tomasz Ludyga
- Department of Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Daria Smyk
- Student Research Circle at the Department of Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Wojciech Smyk
- Student Research Circle at the Department of Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Magdalena Mularska
- Student Research Circle at the Department of Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Patrycja Świderek
- Student Research Circle at the Department of Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Wojciech Majewski
- Department of Radiotherapy, Maria Sklodowska-Curie Institute Oncology Center, Branch in Gliwice, Gliwice, Poland
| | | | - Michael Linnebacher
- Department of General Surgery, Molecular Oncology and Immunotherapy, Rostock University Medical Center, Rostock, Germany
| | - Ewa Obuchowicz
- Department of Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
18
|
Moresi F, Rossetti DV, Vincenzoni F, Simboli GA, La Rocca G, Olivi A, Urbani A, Sabatino G, Desiderio C. Investigating Glioblastoma Multiforme Sub-Proteomes: A Computational Study of CUSA Fluid Proteomic Data. Int J Mol Sci 2022; 23:ijms23042058. [PMID: 35216175 PMCID: PMC8879425 DOI: 10.3390/ijms23042058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/07/2022] [Accepted: 02/10/2022] [Indexed: 02/04/2023] Open
Abstract
Based on our previous proteomic study on Cavitating Ultrasound Aspirator (CUSA) fluid pools of Newly Diagnosed (ND) and Recurrent (R) glioblastomas (GBMs) of tumor core and periphery, as defined by 5-aminolevulinc acid (5-ALA) metabolite fluorescence, this work aims to apply a bioinformatic approach to investigate specifically into three sub-proteomes, i.e., Not Detected in Brain (NB), Cancer Related (CR) and Extracellular Vesicles (EVs) proteins following selected database classification. The study of these yet unexplored specific datasets aims to understand the high infiltration capability and relapse rate that characterizes this aggressive brain cancer. Out of the 587 proteins highly confidently identified in GBM CUSA pools, 53 proteins were classified as NB. Their gene ontology (GO) analysis showed the over-representation of blood coagulation and plasminogen activating cascade pathways, possibly compatible with Blood Brain Barrier damage in tumor disease and surgery bleeding. However, the NB group also included non-blood proteins and, specifically, histones correlated with oncogenesis. Concerning CR proteins, 159 proteins were found in the characterized GBM proteome. Their GO analysis highlighted the over-representation of many pathways, primarily glycolysis. Interestingly, while CR proteins were identified in ND-GBM exclusively in the tumor zones (fluorescence positive core and periphery zones) as predictable, conversely, in R-GBM they were unexpectedly characterized prevalently in the healthy zone (fluorescence negative tumor periphery). Relative to EVs protein classification, 60 proteins were found. EVs are over-released in tumor disease and are important in the transport of biological macromolecules. Furthermore, the presence of EVs in numerous body fluids makes them a possible low-invasive source of brain tumor biomarkers to be investigated. These results give new hints on the molecular features of GBM in trying to understand its aggressive behavior and open to more in-depth investigations to disclose potential disease biomarkers.
Collapse
Affiliation(s)
- Fabiana Moresi
- Department of Neurosurgery, Mater Olbia Hospital, 07026 Olbia, Italy; (F.M.); (G.L.R.); (G.S.)
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.V.); (A.U.)
| | - Diana Valeria Rossetti
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”, Consiglio Nazionale delle Ricerche, 00168 Rome, Italy;
| | - Federica Vincenzoni
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.V.); (A.U.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.A.S.); (A.O.)
| | - Giorgia Antonia Simboli
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.A.S.); (A.O.)
- Institute of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University, 00168 Rome, Italy
| | - Giuseppe La Rocca
- Department of Neurosurgery, Mater Olbia Hospital, 07026 Olbia, Italy; (F.M.); (G.L.R.); (G.S.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.A.S.); (A.O.)
- Institute of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University, 00168 Rome, Italy
| | - Alessandro Olivi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.A.S.); (A.O.)
- Institute of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University, 00168 Rome, Italy
| | - Andrea Urbani
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.V.); (A.U.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.A.S.); (A.O.)
| | - Giovanni Sabatino
- Department of Neurosurgery, Mater Olbia Hospital, 07026 Olbia, Italy; (F.M.); (G.L.R.); (G.S.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.A.S.); (A.O.)
- Institute of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University, 00168 Rome, Italy
| | - Claudia Desiderio
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”, Consiglio Nazionale delle Ricerche, 00168 Rome, Italy;
- Correspondence:
| |
Collapse
|
19
|
Song S, Xiao Z, Dekker FJ, Poelarends GJ, Melgert BN. Macrophage migration inhibitory factor family proteins are multitasking cytokines in tissue injury. Cell Mol Life Sci 2022; 79:105. [PMID: 35091838 PMCID: PMC8799543 DOI: 10.1007/s00018-021-04038-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 02/06/2023]
Abstract
The family of macrophage migration inhibitory factor (MIF) proteins in humans consist of MIF, its functional homolog D-dopachrome tautomerase (D-DT, also known as MIF-2) and the relatively unknown protein named DDT-like (DDTL). MIF is a pleiotropic cytokine with multiple properties in tissue homeostasis and pathology. MIF was initially found to associate with inflammatory responses and therefore established a reputation as a pro-inflammatory cytokine. However, increasing evidence demonstrates that MIF influences many different intra- and extracellular molecular processes important for the maintenance of cellular homeostasis, such as promotion of cellular survival, antioxidant signaling, and wound repair. In contrast, studies on D-DT are scarce and on DDTL almost nonexistent and their functions remain to be further investigated as it is yet unclear how similar they are compared to MIF. Importantly, the many and sometimes opposing functions of MIF suggest that targeting MIF therapeutically should be considered carefully, taking into account timing and severity of tissue injury. In this review, we focus on the latest discoveries regarding the role of MIF family members in tissue injury, inflammation and repair, and highlight the possibilities of interventions with therapeutics targeting or mimicking MIF family proteins.
Collapse
|
20
|
Pediatric glioblastoma: mechanisms of immune evasion and potential therapeutic opportunities. Cancer Immunol Immunother 2022; 71:1813-1822. [PMID: 35020009 DOI: 10.1007/s00262-021-03131-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022]
Abstract
Pediatric glioblastoma is relatively rare compared with its adult counterpart but is associated with a similarly grim prognosis. Available data indicate that pediatric glioblastomas are molecularly distinct from adult tumors, and relatively little is known about the pediatric glioblastoma tumor microenvironment (TME). Cancer immunotherapy has emerged as a new pillar of cancer treatment and is revolutionizing the care of patients with many advanced solid tumors, including melanoma, non-small cell lung cancer, head and neck cancer, and renal cell carcinoma. Unfortunately, attempts to treat adult glioblastoma with current immunotherapies have had limited success to date. Nevertheless, the immune milieu in pediatric glioblastoma is distinct from that found in adult tumors, and evidence suggests that pediatric tumors are less immunosuppressive. As a result, immunotherapies should be specifically evaluated in the pediatric context. The purpose of this review is to explore known and emerging mechanisms of immune evasion in pediatric glioblastoma and highlight potential opportunities for implementing immunotherapy in the treatment of these devastating pediatric brain tumors.
Collapse
|
21
|
Andersen RS, Anand A, Harwood DSL, Kristensen BW. Tumor-Associated Microglia and Macrophages in the Glioblastoma Microenvironment and Their Implications for Therapy. Cancers (Basel) 2021; 13:cancers13174255. [PMID: 34503065 PMCID: PMC8428223 DOI: 10.3390/cancers13174255] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/23/2022] Open
Abstract
Glioblastoma is the most frequent and malignant primary brain tumor. Standard of care includes surgery followed by radiation and temozolomide chemotherapy. Despite treatment, patients have a poor prognosis with a median survival of less than 15 months. The poor prognosis is associated with an increased abundance of tumor-associated microglia and macrophages (TAMs), which are known to play a role in creating a pro-tumorigenic environment and aiding tumor progression. Most treatment strategies are directed against glioblastoma cells; however, accumulating evidence suggests targeting of TAMs as a promising therapeutic strategy. While TAMs are typically dichotomously classified as M1 and M2 phenotypes, recent studies utilizing single cell technologies have identified expression pattern differences, which is beginning to give a deeper understanding of the heterogeneous subpopulations of TAMs in glioblastomas. In this review, we evaluate the role of TAMs in the glioblastoma microenvironment and discuss how their interactions with cancer cells have an extensive impact on glioblastoma progression and treatment resistance. Finally, we summarize the effects and challenges of therapeutic strategies, which specifically aim to target TAMs.
Collapse
Affiliation(s)
- Rikke Sick Andersen
- Department of Pathology, Odense University Hospital, 5000 Odense, Denmark; (R.S.A.); (A.A.)
| | - Atul Anand
- Department of Pathology, Odense University Hospital, 5000 Odense, Denmark; (R.S.A.); (A.A.)
- Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Dylan Scott Lykke Harwood
- Department of Pathology, The Bartholin Institute, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark;
- Department of Clinical Medicine and Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark
| | - Bjarne Winther Kristensen
- Department of Pathology, Odense University Hospital, 5000 Odense, Denmark; (R.S.A.); (A.A.)
- Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
- Department of Pathology, The Bartholin Institute, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark;
- Department of Clinical Medicine and Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark
- Correspondence:
| |
Collapse
|
22
|
Altered Expression of TSPAN32 during B Cell Activation and Systemic Lupus Erythematosus. Genes (Basel) 2021; 12:genes12060931. [PMID: 34207245 PMCID: PMC8234828 DOI: 10.3390/genes12060931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/09/2021] [Accepted: 06/17/2021] [Indexed: 01/02/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic inflammatory disease with various clinical features. Autoreactive B cells play a role in disease pathogenesis, through the production of multiple autoantibodies, which form immune complexes and induce the inflammatory response and tissue damage associated with SLE. Recently, tetraspanins, and in particular, TSPAN32, have been recognized to play a central role in immunity, as they are involved in various biological processes, such as the antigen presentation and the activation of lymphocytes. Evidence suggests that tetraspanins could represent in the future a target for therapeutic purposes in patients suffering from autoimmune/immunoinflammatory disorders. In the present study, by performing in silico analyses of high-throughput data, we evaluated the expression levels of TSPAN32 in B cell activation and investigated its modulation in circulating B cells from SLE patients. Our data show that B cell activation is associated with a significant downregulation of TSPAN32. Additionally, significantly lower levels of TSPAN32 were observed in circulating plasmablasts from SLE patients as compared to healthy donor plasmablasts. In addition, type I interferons (IFNs)-related genes were enriched among the genes negatively correlated to TSPAN32, in SLE plasmablasts. Accordingly, IFN-α is able to induce a dose-dependent downregulation of TSPAN32 in B cells. Overall, the data here presented suggest the potential use of TSPAN32 as a diagnostic marker and therapeutic target for the evaluation and management of humoral immune responses in chronic diseases, such as SLE.
Collapse
|
23
|
Vivarelli S, Falzone L, Basile MS, Candido S, Libra M. Nitric Oxide in Hematological Cancers: Partner or Rival? Antioxid Redox Signal 2021; 34:383-401. [PMID: 32027171 DOI: 10.1089/ars.2019.7958] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Significance: Hematological malignancies represent the fourth most diagnosed cancer. Relapse and acquired resistance to anticancer therapy constitute two actual issues that need to be overcome. Nitric oxide (NO) plays a pivotal role in regulating cancer progression. At present, many studies are attempting to uncover the potentials of modulating NO levels to improve the efficacy of currently available treatments against lymphoma, leukemia, and myeloma. Recent Advances: It is becoming progressively clear that NO modulation may help hematological cancer management, either by targeting directly tumor cells or by driving the immune system to eliminate cancer cells. Critical Issues: NO is a dual molecule that can have a tumor-protecting or stimulating effect, depending on its local concentration. Moreover, NO is able to target a wide range of molecules involved in both cancer genesis and evolution. In this review, an overview of the recent findings regarding the pivotal role played by NO and nitric oxide synthase in cancer progression and anticancer therapy is presented, with particular focus on hematological malignancies. Future Directions: It is critical to establish the cancer-specific function of NO and critically drive its modulation to improve cancer management toward a personalized approach. This has a special importance in hematological tumors, where the urgency of finding eradicative therapies is constant. Antioxid. Redox Signal. 34, 383-401.
Collapse
Affiliation(s)
- Silvia Vivarelli
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Luca Falzone
- Epidemiology Unit, IRCCS Istituto Nazionale Tumori "Fondazione G. Pascale", Napoli, Italy
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Research Centre for Prevention, Diagnosis and Treatment of Cancer, University of Catania, Catania, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Research Centre for Prevention, Diagnosis and Treatment of Cancer, University of Catania, Catania, Italy
| |
Collapse
|
24
|
Abstract
This review considers glioma molecular markers in brain tissues and body fluids, shows the pathways of their formation, and describes traditional methods of analysis. The most important optical properties of glioma markers in the terahertz (THz) frequency range are also presented. New metamaterial-based technologies for molecular marker detection at THz frequencies are discussed. A variety of machine learning methods, which allow the marker detection sensitivity and differentiation of healthy and tumor tissues to be improved with the aid of THz tools, are considered. The actual results on the application of THz techniques in the intraoperative diagnosis of brain gliomas are shown. THz technologies’ potential in molecular marker detection and defining the boundaries of the glioma’s tissue is discussed.
Collapse
|
25
|
Caltabiano R, De Pasquale R, Piombino E, Campo G, Nicoletti F, Cavalli E, Mangano K, Fagone P. Macrophage Migration Inhibitory Factor (MIF) and Its Homologue d-Dopachrome Tautomerase (DDT) Inversely Correlate with Inflammation in Discoid Lupus Erythematosus. Molecules 2021; 26:molecules26010184. [PMID: 33401503 PMCID: PMC7795694 DOI: 10.3390/molecules26010184] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/25/2020] [Accepted: 12/29/2020] [Indexed: 01/12/2023] Open
Abstract
Discoid Lupus Erythematosus (DLE) is a chronic cutaneous disease of unknown etiology and of immunoinflammatory origin that is characterized by inflammatory plaques and may lead to disfiguring scarring and skin atrophy. Current treatments are limited, with a large proportion of patients either poorly or not responsive, which makes DLE an unmet medical need. Macrophage migration inhibitory factor (MIF) is the prototype of a pleiotropic family of cytokine that also includes the recently discovered homologue D-dopachrome tautomerase (DDT) or MIF2. MIF and DDT/MIF-2 exert several biological properties, primarily, but not exclusively of a proinflammatory nature. MIF and DDT have been suggested to play a key role in the pathogenesis of several autoimmune diseases, such as multiple sclerosis and type 1 diabetes, as well as in the development and progression of certain forms of cancers. In the present study, we have performed an immunohistochemistry analysis for the evaluation of MIF in DLE lesions and normal skin. We found high levels of MIF in the basal layer of the epidermis as well as in the cutaneous appendage (eccrine glands and sebocytes) of normal skin. In DLE lesions, we observed a significant negative correlation between the expression of MIF and the severity of inflammation. In addition, we performed an analysis of MIF and DDT expression levels in the skin of DLE patients in a publicly available microarray dataset. Interestingly, while these in silico data only evidenced a trend toward reduced levels of MIF, they demonstrated a significant pattern of expression and correlation of DDT with inflammatory infiltrates in DLE skins. Overall, our data support a protective role for endogenous MIF and possibly DDT in the regulation of homeostasis and inflammation in the skin and open up novel avenues for the treatment of DLE.
Collapse
Affiliation(s)
- Rosario Caltabiano
- Department of Medical, Surgical and Advanced Technologies “G.F. Ingrassia”, University of Catania, Via Santa Sofia, 87, 95123 Catania, Italy; (R.C.); (E.P.)
| | - Rocco De Pasquale
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, Italy;
| | - Eliana Piombino
- Department of Medical, Surgical and Advanced Technologies “G.F. Ingrassia”, University of Catania, Via Santa Sofia, 87, 95123 Catania, Italy; (R.C.); (E.P.)
| | - Giorgia Campo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.C.); (E.C.); (K.M.); (P.F.)
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.C.); (E.C.); (K.M.); (P.F.)
- Correspondence:
| | - Eugenio Cavalli
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.C.); (E.C.); (K.M.); (P.F.)
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.C.); (E.C.); (K.M.); (P.F.)
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.C.); (E.C.); (K.M.); (P.F.)
| |
Collapse
|
26
|
de Azevedo RA, Shoshan E, Whang S, Markel G, Jaiswal AR, Liu A, Curran MA, Travassos LR, Bar-Eli M. MIF inhibition as a strategy for overcoming resistance to immune checkpoint blockade therapy in melanoma. Oncoimmunology 2020; 9:1846915. [PMID: 33344042 PMCID: PMC7733907 DOI: 10.1080/2162402x.2020.1846915] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Immune checkpoint blockade (ICB) has demonstrated an impressive outcome in patients with metastatic melanoma, yet, durable complete response; even with Ipilimumab/Nivolumab combo are under 30%. Primary and acquired resistance in response to ICB is commonly due to a tumor immune escape mechanism dictated by the tumor microenvironment (TME). Macrophage Migratory Inhibition Factor (MIF) has emerged as an immunosuppressive factor secreted in the TME. We have previously demonstrated that blockade of the MIF-CD74 signaling on macrophages and dendritic cells restored the anti-tumor immune response against melanoma. Here, we report that inhibition of the MIF-CD74 axis combined with ipilimumab could render resistant melanoma to better respond to anti-CTLA-4 treatment. We provide evidence that blocking the MIF-CD74 signaling potentiates CD8+ T-cells infiltration and drives pro-inflammatory M1 conversion of macrophages in the TME. Furthermore, MIF inhibition resulted in reprogramming the metabolic pathway by reducing lactate production, HIF-1α and PD-L1 expression in the resistant melanoma cells. Melanoma patient data extracted from the TCGA database supports the hypothesis that high MIF expression strongly correlates with poor response to ICB therapy. Our findings provide a rationale for combining anti-CTLA-4 with MIF inhibitors as a potential strategy to overcome resistance to ICB therapy in melanoma, turning a "cold" tumor into a "hot" one mediated by the activation of innate immunity and reprogramming of tumor metabolism and reduced PD-L1 expression in melanoma cells.
Collapse
Affiliation(s)
- Ricardo A de Azevedo
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Experimental Oncology Unit (UNONEX), Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Einav Shoshan
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shanzhi Whang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gal Markel
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-HaShomer, Israel
| | - Ashvin R Jaiswal
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate School of Biomedical Sciences, MD Anderson Cancer Center UT Health, Houston, TX, USA
| | - Arthur Liu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate School of Biomedical Sciences, MD Anderson Cancer Center UT Health, Houston, TX, USA
| | - Michael A Curran
- Graduate School of Biomedical Sciences, MD Anderson Cancer Center UT Health, Houston, TX, USA
| | - Luiz R Travassos
- Experimental Oncology Unit (UNONEX), Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Menashe Bar-Eli
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
27
|
Lombardo SD, Bramanti A, Ciurleo R, Basile MS, Pennisi M, Bella R, Mangano K, Bramanti P, Nicoletti F, Fagone P. Profiling of inhibitory immune checkpoints in glioblastoma: Potential pathogenetic players. Oncol Lett 2020; 20:332. [PMID: 33123243 PMCID: PMC7583708 DOI: 10.3892/ol.2020.12195] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/06/2020] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma (GBM) represents the most frequent glial tumor, with almost 3 new cases per 100,000 people per year. Despite treatment, the prognosis for GBM patients remains extremely poor, with a median survival of 14.6 months, and a 5-year survival less than 5%. It is generally believed that GBM creates a highly immunosuppressive microenvironment, sustained by the expression of immune-regulatory factors, including inhibitory immune checkpoints, on both infiltrating cells and tumor cells. However, the trials assessing the efficacy of current immune checkpoint inhibitors in GBM are still disappointing. In the present study, the expression levels of several inhibitory immune checkpoints in GBM (CD276, VTCN1, CD47, PVR, TNFRSF14, CD200, LGALS9, NECTIN2 and CD48) were characterized in order to evaluate their potential as prognostic and eventually, therapeutic targets. Among the investigated immune checkpoints, TNFRSF14 and NECTIN2 were identified as the most promising targets in GBM. In particular, a higher TNFRSF14 expression was associated with worse overall survival and disease-free survival, and with a lower Th1 response.
Collapse
Affiliation(s)
- Salvo Danilo Lombardo
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, A-1090 Vienna, Austria
| | | | - Rosella Ciurleo
- IRCCS Centro Neurolesi Bonino Pulejo, I-98124 Messina, Italy
| | | | - Manuela Pennisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| | - Rita Bella
- Department of Medical Sciences, Surgery and Advanced Technologies, University of Catania, I-95123 Catania, Italy
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| | | | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| |
Collapse
|
28
|
Pesce A, Ciurleo R, Bramanti A, Armeli Iapichino EC, Petralia MC, Magro GG, Fagone P, Bramanti P, Nicoletti F, Mangano K. Effects of Combined Admistration of Imatinib and Sorafenib in a Murine Model of Liver Fibrosis. Molecules 2020; 25:molecules25184310. [PMID: 32962198 PMCID: PMC7571085 DOI: 10.3390/molecules25184310] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis is defined as excessive extracellular matrix deposition in the hepatic parenchyma as a consequence of complex interactions among matrix-producing hepatic stellate cells (HSCs) and liver-resident and infiltrating cells. In addition to the liver, the process of fibrosis may represent end-stage disease of several diseases including kidneys, lungs, spleens, heart, muscles and at certain extent, the central nervous system and the peripheral nerves. To date, antifibrotic treatment of fibrosis represents an unconquered area for drug development. The aim of the present study was to test the efficacy of a new drug combination for the treatment of hepatic fibrosis in order to provide a proof-of-concept for the use of therapeutic agents in clinical practice. For this purpose, we have studied the effects of the PDGF inhibitor imatinib and the angiogenesis inhibitor sorafenib, administered alone or in combination, in reducing the progression of the fibrogenetic process in a pre-clinical model of liver damage induced in mice by repeated administration of Concanavalin A (ConA), resembling long-tern autoimmune hepatitis. Our results suggest that treatments with imatinib and sorafenib can modulate potently and, in a superimposable fashion, the fibrinogenic process when administered alone. However, and in agreement with the computational data presently generated, they only exert partial overlapping antifibrotic effects in modulating the main pathways involved in the process of liver fibrosis, without significant additive or synergist effects, when administered in combination.
Collapse
Affiliation(s)
- Antonio Pesce
- Department of Medical and Surgical Sciences and Advanced Technology G.F. Ingrassia, University of Catania, Via Santa Sofia 86, 95123 Catania, Italy; (A.P.); (G.G.M.)
| | - Rosella Ciurleo
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy; (R.C.); (A.B.); (P.B.)
| | - Alessia Bramanti
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy; (R.C.); (A.B.); (P.B.)
| | | | - Maria Cristina Petralia
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.C.P.); (P.F.); (K.M.)
| | - Gaetano Giuseppe Magro
- Department of Medical and Surgical Sciences and Advanced Technology G.F. Ingrassia, University of Catania, Via Santa Sofia 86, 95123 Catania, Italy; (A.P.); (G.G.M.)
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.C.P.); (P.F.); (K.M.)
| | - Placido Bramanti
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy; (R.C.); (A.B.); (P.B.)
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.C.P.); (P.F.); (K.M.)
- Correspondence:
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.C.P.); (P.F.); (K.M.)
| |
Collapse
|
29
|
Zou G, Zhang X, Wang L, Li X, Xie T, Zhao J, Yan J, Wang L, Ye H, Jiao S, Xiang R, Shi Y. Herb-sourced emodin inhibits angiogenesis of breast cancer by targeting VEGFA transcription. Theranostics 2020; 10:6839-6853. [PMID: 32550907 PMCID: PMC7295066 DOI: 10.7150/thno.43622] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 05/06/2020] [Indexed: 02/07/2023] Open
Abstract
Anti-angiogenesis is an important and promising strategy in cancer therapy. However, the current methods using anti-vascular endothelial growth factor A (VEGFA) antibodies or inhibitors targeting VEGFA receptors are not as efficient as expected partly due to their low efficiencies in blocking VEGFA signaling in vivo. Until now, there is still no method to effectively block VEGFA production in cancer cells from the very beginning, i.e., from the transcriptional level. Here, we aimed to find bioactive small molecules to block VEGFA transcription. Methods: We screened our natural compound pool containing 330 small molecules derived from Chinese traditional herbs for small molecules activating the expression of seryl-tRNA synthetase (SerRS), which is a newly identified potent transcriptional repressor of VEGFA, by a cell-based screening system in MDA-MB-231 cell line. The activities of the candidate molecules on regulating SerRS and VEGFA expression were first tested in breast cancer cells. We next investigated the antiangiogenic activity in vivo by testing the effects of candidate drugs on the vascular development in zebrafish and by matrigel plug angiogenesis assay in mice. We further examined the antitumor activities of candidate drugs in two triple-negative breast cancer (TNBC)-bearing mouse models. Furthermore, streptavidin-biotin affinity pull-down assay, coimmunoprecipitation assays, docking analysis and chromatin immunoprecipitation were performed to identify the direct targets of candidate drugs. Results: We identified emodin that could greatly increase SerRS expression in TNBC cells, consequently reducing VEGFA transcription. Emodin potently inhibited vascular development of zebrafish and blocked tumor angiogenesis in TNBC-bearing mice, greatly improving the survival. We also identified nuclear receptor corepressor 2 (NCOR2) to be the direct target of emodin. Once bound by emodin, NCOR2 got released from SerRS promoter, resulting in the activation of SerRS expression and eventually the suppression of VEGFA transcription. Conclusion: We discovered a herb-sourced small molecule emodin with the potential for the therapy of TNBC by targeting transcriptional regulators NCOR2 and SerRS to suppress VEGFA transcription and tumor angiogenesis.
Collapse
Affiliation(s)
- Gengyi Zou
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of Oncology, Chinese PLA General Hospital, Beijing 100853, China
- 2011 Project Collaborative Innovation Center for Biotherapy of Ministry of Education, Tianjin 300071, China
| | - Xiaotong Zhang
- School of Medicine, Nankai University, Tianjin 300071, China
- 2011 Project Collaborative Innovation Center for Biotherapy of Ministry of Education, Tianjin 300071, China
| | - Lun Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Xiyang Li
- School of Medicine, Nankai University, Tianjin 300071, China
- 2011 Project Collaborative Innovation Center for Biotherapy of Ministry of Education, Tianjin 300071, China
| | - Tianyu Xie
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Jin Zhao
- School of Medicine, Nankai University, Tianjin 300071, China
- 2011 Project Collaborative Innovation Center for Biotherapy of Ministry of Education, Tianjin 300071, China
| | - Jie Yan
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Longlong Wang
- School of Medicine, Nankai University, Tianjin 300071, China
- 2011 Project Collaborative Innovation Center for Biotherapy of Ministry of Education, Tianjin 300071, China
| | - Haoyu Ye
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Shunchang Jiao
- Department of Oncology, Chinese PLA General Hospital, Beijing 100853, China
| | - Rong Xiang
- School of Medicine, Nankai University, Tianjin 300071, China
- 2011 Project Collaborative Innovation Center for Biotherapy of Ministry of Education, Tianjin 300071, China
| | - Yi Shi
- School of Medicine, Nankai University, Tianjin 300071, China
- 2011 Project Collaborative Innovation Center for Biotherapy of Ministry of Education, Tianjin 300071, China
| |
Collapse
|
30
|
Kang J, Feng D, Yang F, Tian X, Han W, Jia H. Comparison of rapamycin and methylprednisolone for treating inflammatory muscle disease in a murine model of experimental autoimmune myositis. Exp Ther Med 2020; 20:219-226. [PMID: 32536994 PMCID: PMC7291653 DOI: 10.3892/etm.2020.8716] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 12/09/2019] [Indexed: 01/03/2023] Open
Abstract
Idiopathic inflammatory myopathies (IIMs) are a group of autoimmune inflammatory muscle diseases. Rapamycin has been shown to ameliorate inflammation and improve muscle function in a mouse model of experimental autoimmune myositis (EAM). In the present study, the therapeutic effect of rapamycin was compared with methylprednisolone (MP) on EAM. Mice were injected with myosin for 10 days to induce EAM and were subsequently treated with rapamycin (1.5 mg/kg), MP (40 mg/kg) or placebo (DMSO) for 14 days. The rapamycin-treated group exhibited significantly decreased severe inflammation and improved muscle strength compared with the MP-treated group. The plasma transforming growth factor-β (TGF-β) concentration in the rapamycin-treated group was significantly higher compared with the placebo group. However, both treatment groups exhibited significantly lower plasma interleukin-10 levels compared with the placebo group. Moreover, splenic regulatory T cell frequency in both the rapamycin- and MP-treated animals was significantly lower than that in the animals of the placebo group. Rapamycin showed better immune suppressive effects than MP in this model of EAM, and these effects were likely to be mediated by the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Juan Kang
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xincheng, Xi'an, Shaanxi 710032, P.R. China
| | - Dongyun Feng
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xincheng, Xi'an, Shaanxi 710032, P.R. China
| | - Feng Yang
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xincheng, Xi'an, Shaanxi 710032, P.R. China
| | - Xiaojia Tian
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xincheng, Xi'an, Shaanxi 710032, P.R. China
| | - Wenjuan Han
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xincheng, Xi'an, Shaanxi 710032, P.R. China
| | - Hongge Jia
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xincheng, Xi'an, Shaanxi 710032, P.R. China.,Department of Neurology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong 518034, P.R. China
| |
Collapse
|
31
|
The Dichotomic Role of Macrophage Migration Inhibitory Factor in Neurodegeneration. Int J Mol Sci 2020; 21:ijms21083023. [PMID: 32344747 PMCID: PMC7216212 DOI: 10.3390/ijms21083023] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 12/12/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine expressed by different cell types and exerting multiple biological functions. It has been shown that MIF may be involved in several disorders, including neurodegenerative disorders such as amyotrophic lateral sclerosis (ALS), Parkinson disease (PD), and Huntington disease (HD), that represent an unmet medical need. Therefore, further studies are needed to identify novel pathogenetic mechanisms that may translate into tailored therapeutic approaches so to improve patients’ survival and quality of life. Here, we reviewed the preclinical and clinical studies investigating the role of MIF in ALS, PD, and HD. The emerging results suggest that MIF might play a dichotomic role in these disorders, exerting a protective action in ALS, a pathogenetic action in HD, and a yet undefined and debated role in PD. The better understanding of the role of MIF in these diseases could allow its use as a novel diagnostic and therapeutic tool for the monitoring and treatment of the patients and for eventual biomarker-driven therapeutic approaches.
Collapse
|
32
|
Michaličková D, Hrnčíř T, Canová NK, Slanař O. Targeting Keap1/Nrf2/ARE signaling pathway in multiple sclerosis. Eur J Pharmacol 2020; 873:172973. [DOI: 10.1016/j.ejphar.2020.172973] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/08/2020] [Accepted: 01/28/2020] [Indexed: 12/29/2022]
|
33
|
Cavalli E, Battaglia G, Basile MS, Bruno V, Petralia MC, Lombardo SD, Pennisi M, Kalfin R, Tancheva L, Fagone P, Nicoletti F, Mangano K. Exploratory Analysis of iPSCS-Derived Neuronal Cells as Predictors of Diagnosis and Treatment of Alzheimer Disease. Brain Sci 2020; 10:brainsci10030166. [PMID: 32183090 PMCID: PMC7139610 DOI: 10.3390/brainsci10030166] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/28/2020] [Accepted: 03/11/2020] [Indexed: 12/13/2022] Open
Abstract
Alzheimer’s disease (AD) represents the most common neurodegenerative disorder, with 47 million affected people worldwide. Current treatment strategies are aimed at reducing the symptoms and do slow down the progression of the disease, but inevitably fail in the long-term. Induced pluripotent stem cells (iPSCs)-derived neuronal cells from AD patients have proven to be a reliable model for AD pathogenesis. Here, we have conducted an in silico analysis aimed at identifying pathogenic gene-expression profiles and novel drug candidates. The GSE117589 microarray dataset was used for the identification of Differentially Expressed Genes (DEGs) between iPSC-derived neuronal progenitor (NP) cells and neurons from AD patients and healthy donors. The Discriminant Analysis Module (DAM) algorithm was used for the identification of biomarkers of disease. Drugs with anti-signature gene perturbation profiles were identified using the L1000FWD software. DAM analysis was used to identify a list of potential biomarkers among the DEGs, able to discriminate AD patients from healthy people. Finally, anti-signature perturbation analysis identified potential anti-AD drugs. This study set the basis for the investigation of potential novel pharmacological strategies for AD. Furthermore, a subset of genes for the early diagnosis of AD is proposed.
Collapse
Affiliation(s)
- Eugenio Cavalli
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (E.C.); (M.S.B.); (S.D.L.); (M.P.); (F.N.); (K.M.)
| | - Giuseppe Battaglia
- University Sapienza, Piazzale A. Moro, 5, 00185 Roma, Italy; (G.B.); (V.B.)
- IRCCS Neuromed, Località Camerelle, 86077 Pozzilli (IS), Italy
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (E.C.); (M.S.B.); (S.D.L.); (M.P.); (F.N.); (K.M.)
| | - Valeria Bruno
- University Sapienza, Piazzale A. Moro, 5, 00185 Roma, Italy; (G.B.); (V.B.)
- IRCCS Neuromed, Località Camerelle, 86077 Pozzilli (IS), Italy
| | | | - Salvo Danilo Lombardo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (E.C.); (M.S.B.); (S.D.L.); (M.P.); (F.N.); (K.M.)
| | - Manuela Pennisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (E.C.); (M.S.B.); (S.D.L.); (M.P.); (F.N.); (K.M.)
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Block 23, 1113 Sofia, Bulgaria; (R.K.); (L.T.)
| | - Lyubka Tancheva
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Block 23, 1113 Sofia, Bulgaria; (R.K.); (L.T.)
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (E.C.); (M.S.B.); (S.D.L.); (M.P.); (F.N.); (K.M.)
- Correspondence: ; Tel.: +39-095-478-1284
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (E.C.); (M.S.B.); (S.D.L.); (M.P.); (F.N.); (K.M.)
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (E.C.); (M.S.B.); (S.D.L.); (M.P.); (F.N.); (K.M.)
| |
Collapse
|
34
|
Emerging Role of the Macrophage Migration Inhibitory Factor Family of Cytokines in Neuroblastoma. Pathogenic Effectors and Novel Therapeutic Targets? Molecules 2020; 25:molecules25051194. [PMID: 32155795 PMCID: PMC7179464 DOI: 10.3390/molecules25051194] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 12/17/2022] Open
Abstract
Neuroblastoma (NB) is the most frequent extracranial pediatric tumor. Despite the current available multiple therapeutic options, the prognosis for high-risk NB patients remains unsatisfactory and makes the disease a clear unmet medical need. Thus, more tailored therapeutic approaches are warranted to improve both the quality of life and the survival of the patients. Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine that plays a key role in several diseases, including cancer. Preclinical and clinical studies in NB patients convergently indicate that MIF exerts pro-tumorigenic properties in NB. MIF is upregulated in NB tumor tissues and cell lines and it contributes to NB aggressiveness and immune-escape. To date, there are only a few data about the role of the second member of the MIF family, the MIF homolog d-dopachrome tautomerase (DDT), in NB. Here, we review the preclinical and clinical studies on the role of the MIF family of cytokines in NB and suggest that MIF and possibly DDT inhibitors may be promising novel prognostic and therapeutic targets in NB management.
Collapse
|
35
|
Pathogenic contribution of the Macrophage migration inhibitory factor family to major depressive disorder and emerging tailored therapeutic approaches. J Affect Disord 2020; 263:15-24. [PMID: 31818772 DOI: 10.1016/j.jad.2019.11.127] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/28/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Immunoinflammatory disorders are often accompanied by depression. Here, we review the available preclinical and clinical studies suggesting a role for the pro-inflammatory cytokine Macrophage migration inhibitory factor (MIF) and the second member of the MIF family, D-dopachrome tautomerase (D-DT; DDT), in the pathogenesis of Major Depressive Disorders (MDD). METHODS We prepared a narrative review from a search on PubMed of studies pertaining to MDD and MIF, as for October 2019. Both humans and animal studies haves been considered. RESULTS Preclinical data show conflicting results on the role of endogenous MIF and DDT in depression. In contrast, several human studies show that circulating MIF levels tend to increase during the course of MDD. Higher levels of inflammatory biomarkers have also been associated with poorer responses to antidepressants and the levels of MIF significantly decrease after treatment, despite this may not be necessarily associated to an improvement in psychiatric symptoms. LIMITATIONS This is a narrative and not a systematic review of the literature on the involvement of MIF in MDD. We have highlighted studies performed in humans and in animal models, irrespective of population size and methodological approach. CONCLUSIONS This review highlights a role of MIF, and possibly DDT, in the pathogenesis of MDD. Whilst studies in animal models are discordant, the studies in patients with MDD convergently suggest that MIF plays a role in induction and maintenance of the disease. Additional studies are also needed on DDT that often displays synergistic function with MIF and their receptors.
Collapse
|
36
|
Mangano K, Lanteri R, Basile MS, Bellavia N, Latino R, Messina D, Fagone P, Colletti G, Nania R, Caltabiano R, Di Marco R, Di Cataldo A. Effects of GIT-27NO, a NO-donating compound, on hepatic ischemia/reperfusion injury. Int J Immunopathol Pharmacol 2020; 33:2058738419862736. [PMID: 31298048 PMCID: PMC6628530 DOI: 10.1177/2058738419862736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Hepatic ischemia/reperfusion injury (IRI) is a clinical condition that may lead
to cellular injury and organ dysfunction that can be observed in different
conditions, such as trauma, shock, liver resection, and transplantation.
Moderate levels of nitric oxide (NO) produced by the endothelial isoform of the
NO synthase protect against liver IRI. GIT-27NO is a NO-derivative of the
toll-like receptor 4 antagonist VGX-1027 that has been shown to possess both
antineoplastic and immunomodulatory properties in vitro and in vivo. In this
study, we have investigated the effects of this compound in vitro, in a model of
oxidative stress induced in HepG2 cells by hydrogen peroxide
(H2O2), and in vivo, in a rat model of IRI of the
liver. GIT-27NO significantly counteracted the toxic effects induced by the
H2O2 on the HepG2 cells and in vivo, GIT-27NO reduced
the transaminase levels and the histological liver injury by reducing necrotic
areas with preservation of viable tissue. These effects were almost similar to
that of the positive control drug dimethyl fumarate. These data suggest that the
beneficial effect of GIT-27NO in the hepatic IRI can be secondary to
anti-oxidative effects and hepatocyte necrosis reduction probably mediated by NO
release.
Collapse
Affiliation(s)
- Katia Mangano
- 1 Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Raffaele Lanteri
- 2 Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Maria Sofia Basile
- 1 Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Noemi Bellavia
- 2 Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Rosalia Latino
- 2 Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Domenico Messina
- 1 Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Paolo Fagone
- 1 Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuseppe Colletti
- 1 Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Riccardo Nania
- 1 Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosario Caltabiano
- 3 Department of Surgical Sciences, Organ Transplantation and Advanced Technologies, G.F. Ingrassia, University of Catania, Catania, Italy
| | - Roberto Di Marco
- 4 Department of Medicine and Health Sciences "Vincenzo Tiberio," University of Molise, Campobasso, Italy
| | - Antonio Di Cataldo
- 2 Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| |
Collapse
|
37
|
Basile MS, Mazzon E, Mangano K, Pennisi M, Petralia MC, Lombardo SD, Nicoletti F, Fagone P, Cavalli E. Impaired Expression of Tetraspanin 32 (TSPAN32) in Memory T Cells of Patients with Multiple Sclerosis. Brain Sci 2020; 10:brainsci10010052. [PMID: 31963428 PMCID: PMC7016636 DOI: 10.3390/brainsci10010052] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/08/2020] [Accepted: 01/14/2020] [Indexed: 02/08/2023] Open
Abstract
Tetraspanins are a conserved family of proteins involved in a number of biological processes. We have previously shown that Tetraspanin-32 (TSPAN32) is significantly downregulated upon activation of T helper cells via anti-CD3/CD28 stimulation. On the other hand, TSPAN32 is marginally modulated in activated Treg cells. A role for TSPAN32 in controlling the development of autoimmune responses is consistent with our observation that encephalitogenic T cells from myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) mice exhibit significantly lower levels of TSPAN32 as compared to naïve T cells. In the present study, by making use of ex vivo and in silico analysis, we aimed to better characterize the pathophysiological and diagnostic/prognostic role of TSPAN32 in T cell immunity and in multiple sclerosis (MS). We first show that TSPAN32 is significantly downregulated in memory T cells as compared to naïve T cells, and that it is further diminished upon ex vivo restimulation. Accordingly, following antigenic stimulation, myelin-specific memory T cells from MS patients showed significantly lower expression of TSPAN32 as compared to memory T cells from healthy donors (HD). The expression levels of TSPAN32 was significantly downregulated in peripheral blood mononuclear cells (PBMCs) from drug-naïve MS patients as compared to HD, irrespective of the disease state. Finally, when comparing patients undergoing early relapses in comparison to patients with longer stable disease, moderate but significantly lower levels of TSPAN32 expression were observed in PBMCs from the former group. Our data suggest a role for TSPAN32 in the immune responses underlying the pathophysiology of MS and represent a proof-of-concept for additional studies aiming at dissecting the eventual contribution of TSPAN32 in other autoimmune diseases and its possible use of TSPAN32 as a diagnostic factor and therapeutic target.
Collapse
Affiliation(s)
- Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (M.S.B.); (K.M.); (M.P.); (S.D.L.); (F.N.)
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (E.M.); (M.C.P.); (E.C.)
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (M.S.B.); (K.M.); (M.P.); (S.D.L.); (F.N.)
| | - Manuela Pennisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (M.S.B.); (K.M.); (M.P.); (S.D.L.); (F.N.)
| | - Maria Cristina Petralia
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (E.M.); (M.C.P.); (E.C.)
| | - Salvo Danilo Lombardo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (M.S.B.); (K.M.); (M.P.); (S.D.L.); (F.N.)
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (M.S.B.); (K.M.); (M.P.); (S.D.L.); (F.N.)
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (M.S.B.); (K.M.); (M.P.); (S.D.L.); (F.N.)
- Correspondence:
| | - Eugenio Cavalli
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (E.M.); (M.C.P.); (E.C.)
| |
Collapse
|
38
|
The Role of Macrophage Migration Inhibitory Factor in Alzheimer's Disease: Conventionally Pathogenetic or Unconventionally Protective? Molecules 2020; 25:molecules25020291. [PMID: 31936865 PMCID: PMC7024279 DOI: 10.3390/molecules25020291] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/03/2020] [Accepted: 01/09/2020] [Indexed: 02/06/2023] Open
Abstract
Recent preclinical and clinical observations have offered relevant insights on the etiopathogenesis of late onset Alzheimer′s disease (AD) and upregulated immunoinflammatory events have been described as underlying mechanisms involved in the development of AD. Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine produced by several cells of the innate and adaptive immune system, as well as non-immune cells. In the present review, we highlight experimental, genetic, and clinical studies on MIF in rodent models of AD and AD patients, and we discuss emerging therapeutic opportunities for tailored modulation of the activity of MIF, that may potentially be applied to AD patients. Dismantling the exact role of MIF and its receptors in AD may offer novel diagnostic and therapeutic opportunities in AD.
Collapse
|
39
|
Petralia MC, Mazzon E, Mangano K, Fagone P, Di Marco R, Falzone L, Basile MS, Nicoletti F, Cavalli E. Transcriptomic analysis reveals moderate modulation of macrophage migration inhibitory factor superfamily genes in alcohol use disorders. Exp Ther Med 2020; 19:1755-1762. [PMID: 32104230 PMCID: PMC7026954 DOI: 10.3892/etm.2020.8410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/14/2019] [Indexed: 12/20/2022] Open
Abstract
Alcohol use disorder (AUD) is a primary, chronic and relapsing disease of brain reward, motivation and memory, which is associated with several comorbidities, including major depression and post-traumatic stress disorder. It has been revealed that Ibudilast (IBUD), a dual inhibitor of phosphodiesterase-4 and −10 and of macrophage migration inhibitory factor (MIF), exerts beneficial effects on AUD in rodent models and human patients. Therefore, IBUD has attracted increasing interest, with research focusing on the elucidation of the pathogenic role of MIF and its homologue, D-dopachrome tautomerase (DDT), in the pathogenesis and maintenance of AUD. By using DNA microarray analysis, the current study performed a transcriptomic expression analysis of MIF, DDT and their co-receptors, including CD74, C-X-C chemokine receptor (CXCR)2, CXCR4 and CXCR7 in patients with AUD. The results revealed that the transcriptomic levels of MIF, DDT and their receptors were superimposable in the prefrontal cortex of rodents and patients with AUD and human patients. Furthermore, peripheral blood cells from heavy drinkers exhibited a moderate increase in MIF and DDT levels, both at the baseline and following exposure to alcohol-associated cues, based on individual situations that included alcohol-related stimuli resulting in subsequent alcohol use (buying alcohol and being at a bar, watching others drink alcohol). Considering the overlapping effects of MIF and DDT, the inverse Fisher's χ2 test was performed on unadjusted P-values to evaluate the combined effect of MIF and DDT. The results revealed a significant increase in these cytokines in heavy drinkers compared with controls (moderate drinkers). To the best of our knowledge, the present study demonstrated for the first time that MIF and DDT expression was upregulated in the blood of patients with AUD. These results therefore warrant further study to evaluate the role of MIF and DDT in the development and maintenance of AUD, to evaluate their use as biomarkers to predict the psychotherapeutic and pharmacological response of patients with AUD and for use as therapeutic targets.
Collapse
Affiliation(s)
- Maria Cristina Petralia
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| | - Emanuela Mazzon
- IRCCS (Scientific Institute for Research, Hospitalization and Healthcare) Centro Neurolesi 'Bonino-Pulejo', I-98124 Messina, Italy
| | - Katia Mangano
- IRCCS (Scientific Institute for Research, Hospitalization and Healthcare) Centro Neurolesi 'Bonino-Pulejo', I-98124 Messina, Italy
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| | - Roberto Di Marco
- Department of Medicine and Health Sciences 'Vincenzo Tiberio', University of Molise, I-86100 Campobasso, Italy
| | - Luca Falzone
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| | - Eugenio Cavalli
- IRCCS (Scientific Institute for Research, Hospitalization and Healthcare) Centro Neurolesi 'Bonino-Pulejo', I-98124 Messina, Italy
| |
Collapse
|
40
|
Wu Q, Chen X, He Q, Lang L, Xu P, Wang P, Lee SC. Resveratrol attenuates diabetes-associated cell centrosome amplification via inhibiting the PKCα-p38 to c-myc/c-jun pathway. Acta Biochim Biophys Sin (Shanghai) 2020; 52:72-83. [PMID: 31844893 DOI: 10.1093/abbs/gmz142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 09/06/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023] Open
Abstract
Type 2 diabetes increases the risk for cancer. Centrosome amplification can initiate tumorigenesis. We have described that type 2 diabetes increases the centrosome amplification of peripheral blood mononuclear cells, with high glucose, insulin, and palmitic acid as the triggers, which suggests that centrosome amplification is a candidate biological mechanism linking diabetes to cancer. In this study, we aimed to further investigate the signaling pathways of the diabetes-associated centrosome amplification and to examine whether and how resveratrol inhibits the centrosome amplification. The results showed that treatment with high glucose, insulin, and palmitic acid, alone or in combination, could increase the protein levels of phospho-protein kinase C alpha (p-PKCα), phospho-p38 mitogen-activated protein kinases (p-p38), c-myc, and c-jun, as well as the mRNA levels of c-myc and c-jun. PKCα inhibitor could inhibit the treatment-induced increase in the protein levels of p-p38, c-myc, and c-jun. Inhibitor or siRNA of p38 was also able to inhibit the treatment-induced increase in the levels of p-p38, c-myc, and c-jun. Meanwhile, knockdown of c-myc or c-jun did not alter the treatment-induced increase in the phosphorylation of PKCα or p38. Importantly, inhibition of the phosphorylation of PKCα or p38 and knockdown of c-myc or c-jun could attenuate the centrosome amplification. In diabetic mice, the levels of p-PKCα, p-p38, c-myc, and c-jun were all increased in the colon tissues. Interestingly, resveratrol, but not metformin, was able to attenuate the treatment-induced increase in the levels of p-PKCα, p-p38, c-myc, and c-jun, as well as the centrosome amplification. In conclusion, our results suggest that PKCα-p38 to c-myc/c-jun is the signaling pathway of the diabetes-associated centrosome amplification, and resveratrol attenuates the centrosome amplification by inhibiting this signaling pathway.
Collapse
Affiliation(s)
- Qigui Wu
- School of Life Sciences, Shanxi University, Taiyuan 030006, China, and
| | - Xiaoyu Chen
- School of Life Sciences, Shanxi University, Taiyuan 030006, China, and
| | - Qinju He
- School of Life Sciences, Shanxi University, Taiyuan 030006, China, and
| | - Lang Lang
- School of Life Sciences, Shanxi University, Taiyuan 030006, China, and
| | - Peng Xu
- School of Life Sciences, Shanxi University, Taiyuan 030006, China, and
| | - Pu Wang
- School of Life Sciences, Shanxi University, Taiyuan 030006, China, and
| | - Shao Chin Lee
- School of Life Sciences, Shanxi University, Taiyuan 030006, China, and
- School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, China
| |
Collapse
|
41
|
Lopes-Coelho F, Silva F, Gouveia-Fernandes S, Martins C, Lopes N, Domingues G, Brito C, Almeida AM, Pereira SA, Serpa J. Monocytes as Endothelial Progenitor Cells (EPCs), Another Brick in the Wall to Disentangle Tumor Angiogenesis. Cells 2020; 9:cells9010107. [PMID: 31906296 PMCID: PMC7016533 DOI: 10.3390/cells9010107] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/22/2019] [Accepted: 12/30/2019] [Indexed: 12/12/2022] Open
Abstract
Bone marrow contains endothelial progenitor cells (EPCs) that, upon pro-angiogenic stimuli, migrate and differentiate into endothelial cells (ECs) and contribute to re-endothelialization and neo-vascularization. There are currently no reliable markers to characterize EPCs, leading to their inaccurate identification. In the past, we showed that, in a panel of tumors, some cells on the vessel wall co-expressed CD14 (monocytic marker) and CD31 (EC marker), indicating a putative differentiation route of monocytes into ECs. Herein, we disclosed monocytes as potential EPCs, using in vitro and in vivo models, and also addressed the cancer context. Monocytes acquired the capacity to express ECs markers and were able to be incorporated into blood vessels, contributing to cancer progression, by being incorporated in tumor neo-vasculature. Reactive oxygen species (ROS) push monocytes to EC differentiation, and this phenotype is reverted by cysteine (a scavenger and precursor of glutathione), which indicates that angiogenesis is controlled by the interplay between the oxidative stress and the scavenging capacity of the tumor microenvironment.
Collapse
Affiliation(s)
- Filipa Lopes-Coelho
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Fernanda Silva
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Sofia Gouveia-Fernandes
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Carmo Martins
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Nuno Lopes
- Instituto de Biologia Experimental e Tecnológica, Avenida da República, Estação Agronómica, 2780-157 Oeiras, Portugal; (N.L.); (C.B.)
| | - Germana Domingues
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Catarina Brito
- Instituto de Biologia Experimental e Tecnológica, Avenida da República, Estação Agronómica, 2780-157 Oeiras, Portugal; (N.L.); (C.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - António M Almeida
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
- Hospital da Luz, Av. Lusíada 100, 1500-650 Lisboa, Portugal
| | - Sofia A Pereira
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
| | - Jacinta Serpa
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
- Correspondence: ; Tel.: +350-217-229-800; Fax: +351-217-248-756
| |
Collapse
|
42
|
Cavalli E, Mazzon E, Basile MS, Mammana S, Pennisi M, Fagone P, Kalfin R, Martinovic V, Ivanovic J, Andabaka M, Mesaros S, Pekmezovic T, Drulovic J, Nicoletti F, Petralia MC. In Silico and In Vivo Analysis of IL37 in Multiple Sclerosis Reveals Its Probable Homeostatic Role on the Clinical Activity, Disability, and Treatment with Fingolimod. Molecules 2019; 25:molecules25010020. [PMID: 31861585 PMCID: PMC6982851 DOI: 10.3390/molecules25010020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/12/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022] Open
Abstract
We evaluated the in silico expression and circulating levels of interleukin (IL)37 in patients with different forms of multiple sclerosis (MS) and also upon treatment with different disease-modifying drugs. The combined interpretation of the resulting data strengthens and extends the current emerging concept that endogenous IL37 plays an important role in determining onset and progression of MS. The in silico analysis revealed that production of IL37 from cluster of differentiation (CD)4+ T cells from MS patients was reduced in vitro as compared to healthy controls. The analysis of the datasets also demonstrated that “higher” levels of IL37 production from PBMC entailed significant protection from MS relapses. In addition, the in vivo part of the study showed that IL37 was selectively augmented in the sera of MS patients during a relapse and that treatment with the high potency disease-modifying drug fingolimod significantly increased the frequency of patients with circulating blood levels of IL37 (6/9, 66%) as compared to patients receiving no treatment (n = 48) or platform therapy (n = 59) who had levels of IL37 below the limit of the sensitivity of the assay. This finding therefore anticipates that fingolimod may at least partially exert its beneficial effects in MS by upregulating the production of IL37.
Collapse
Affiliation(s)
- Eugenio Cavalli
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (E.C.); (E.M.); (S.M.); (M.C.P.)
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (E.C.); (E.M.); (S.M.); (M.C.P.)
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (M.S.B.); (M.P.); (P.F.)
| | - Santa Mammana
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (E.C.); (E.M.); (S.M.); (M.C.P.)
| | - Manuela Pennisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (M.S.B.); (M.P.); (P.F.)
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (M.S.B.); (M.P.); (P.F.)
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Block 23 1113 Sofia, Bulgaria;
| | - Vanja Martinovic
- Clinic of Neurology, Clinical Center of Serbia, Dr Subotica 6, 11000 Belgrade, Serbia; (V.M.); (J.I.); (M.A.); (S.M.); (T.P.); (J.D.)
| | - Jovana Ivanovic
- Clinic of Neurology, Clinical Center of Serbia, Dr Subotica 6, 11000 Belgrade, Serbia; (V.M.); (J.I.); (M.A.); (S.M.); (T.P.); (J.D.)
| | - Marko Andabaka
- Clinic of Neurology, Clinical Center of Serbia, Dr Subotica 6, 11000 Belgrade, Serbia; (V.M.); (J.I.); (M.A.); (S.M.); (T.P.); (J.D.)
| | - Sarlota Mesaros
- Clinic of Neurology, Clinical Center of Serbia, Dr Subotica 6, 11000 Belgrade, Serbia; (V.M.); (J.I.); (M.A.); (S.M.); (T.P.); (J.D.)
| | - Tatjana Pekmezovic
- Clinic of Neurology, Clinical Center of Serbia, Dr Subotica 6, 11000 Belgrade, Serbia; (V.M.); (J.I.); (M.A.); (S.M.); (T.P.); (J.D.)
| | - Jelena Drulovic
- Clinic of Neurology, Clinical Center of Serbia, Dr Subotica 6, 11000 Belgrade, Serbia; (V.M.); (J.I.); (M.A.); (S.M.); (T.P.); (J.D.)
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (M.S.B.); (M.P.); (P.F.)
- Correspondence: ; Tel.: +39-095-478-1270
| | - Maria Cristina Petralia
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (E.C.); (E.M.); (S.M.); (M.C.P.)
| |
Collapse
|
43
|
Petralia MC, Mazzon E, Basile MS, Cutuli M, Di Marco R, Scandurra F, Saraceno A, Fagone P, Nicoletti F, Mangano K. Effects of Treatment with the Hypomethylating Agent 5-aza-2'-deoxycytidine in Murine Type II Collagen-Induced Arthritis. Pharmaceuticals (Basel) 2019; 12:ph12040174. [PMID: 31783688 PMCID: PMC6958460 DOI: 10.3390/ph12040174] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 11/26/2019] [Accepted: 11/26/2019] [Indexed: 02/06/2023] Open
Abstract
The emerging role of epigenetics in the pathogenesis of autoimmune diseases has recently attracted much interest on the possible use of epigenetic modulators for the prevention and treatment of these diseases. In particular, we and others have shown that drugs that inhibit DNA methylation, such as azacitidine (AZA) and decitabine (DAC), already used for the treatment of acute myeloid leukemia, exert powerful beneficial effects in rodent models of type 1 diabetes, multiple sclerosis, and Guillain Barrè syndrome. Along this line of research, we have presently studied the effects of DAC in a murine model of rheumatoid arthritis induced by type II collagen and have demonstrated that DAC administration was associated with a significant amelioration of the clinical condition, along with in vivo and ex vivo modification of the immunological profile of the so-treated mice, that exhibited a diminished production of Th1 and Th17 pro-inflammatory cytokines and reduction of anti-type II collagen autoantibodies.
Collapse
Affiliation(s)
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy; (M.C.P.); (E.M.)
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.S.B.); (F.S.); (A.S.); (P.F.); (K.M.)
| | - Marco Cutuli
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, 86100 Campobasso, Italy; (M.C.); (R.D.M.)
| | - Roberto Di Marco
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, 86100 Campobasso, Italy; (M.C.); (R.D.M.)
| | - Fabiola Scandurra
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.S.B.); (F.S.); (A.S.); (P.F.); (K.M.)
| | - Andrea Saraceno
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.S.B.); (F.S.); (A.S.); (P.F.); (K.M.)
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.S.B.); (F.S.); (A.S.); (P.F.); (K.M.)
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.S.B.); (F.S.); (A.S.); (P.F.); (K.M.)
- Correspondence: ; Tel.: +39-095-478-1270
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.S.B.); (F.S.); (A.S.); (P.F.); (K.M.)
| |
Collapse
|
44
|
Transcriptomic Analysis Reveals Involvement of the Macrophage Migration Inhibitory Factor Gene Network in Duchenne Muscular Dystrophy. Genes (Basel) 2019; 10:genes10110939. [PMID: 31752120 PMCID: PMC6896047 DOI: 10.3390/genes10110939] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/11/2019] [Accepted: 11/15/2019] [Indexed: 01/04/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive hereditary muscular disease with X-linked recessive inheritance, that leads patients to premature death. The loss of dystrophin determines membrane instability, causing cell damage and inflammatory response. Macrophage migration inhibitory factor (MIF) is a cytokine that exerts pleiotropic properties and is implicated in the pathogenesis of a variety of diseases. Recently, converging data from independent studies have pointed to a possible role of MIF in dystrophic muscle disorders, including DMD. In the present study, we have investigated the modulation of MIF and MIF-related genes in degenerative muscle disorders, by making use of publicly available whole-genome expression datasets. We show here a significant enrichment of MIF and related genes in muscle samples from DMD patients, as well as from patients suffering from Becker’s disease and limb-girdle muscular dystrophy type 2B. On the other hand, transcriptomic analysis of in vitro differentiated myotubes from healthy controls and DMD patients revealed no significant alteration in the expression levels of MIF-related genes. Finally, by analyzing DMD samples as a time series, we show that the modulation of the genes belonging to the MIF network is an early event in the DMD muscle and does not change with the increasing age of the patients, Overall, our analysis suggests that MIF may play a role in vivo during muscle degeneration, likely promoting inflammation and local microenvironment reaction.
Collapse
|
45
|
Basile MS, Mazzon E, Fagone P, Longo A, Russo A, Fallico M, Bonfiglio V, Nicoletti F, Avitabile T, Reibaldi M. Immunobiology of Uveal Melanoma: State of the Art and Therapeutic Targets. Front Oncol 2019; 9:1145. [PMID: 31750244 PMCID: PMC6848265 DOI: 10.3389/fonc.2019.01145] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 10/15/2019] [Indexed: 12/16/2022] Open
Abstract
Uveal Melanoma (UM) represents the most common primary intraocular malignant tumor in adults. Although it originates from melanocytes as cutaneous melanoma, it shows significant clinical and biological differences with the latter, including high resistance to immune therapy. Indeed, UM can evade immune surveillance via multiple mechanisms, such as the expression of inhibitory checkpoints (e.g., PD-L1, CD47, CD200) and the production of IDO-1 and soluble FasL, among others. More in-depth understanding of these mechanisms will suggest potential targets for the design of novel and more effective management strategies for UM patients.
Collapse
Affiliation(s)
- Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, Messina, Italy
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Antonio Longo
- Department of Ophthalmology, University of Catania, Catania, Italy
| | - Andrea Russo
- Department of Ophthalmology, University of Catania, Catania, Italy
| | - Matteo Fallico
- Department of Ophthalmology, University of Catania, Catania, Italy
| | | | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Michele Reibaldi
- Department of Ophthalmology, University of Catania, Catania, Italy
| |
Collapse
|
46
|
Cavalli E, Mazzon E, Mammana S, Basile MS, Lombardo SD, Mangano K, Bramanti P, Nicoletti F, Fagone P, Petralia MC. Overexpression of Macrophage Migration Inhibitory Factor and Its Homologue D-Dopachrome Tautomerase as Negative Prognostic Factor in Neuroblastoma. Brain Sci 2019; 9:brainsci9100284. [PMID: 31635049 PMCID: PMC6826588 DOI: 10.3390/brainsci9100284] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/01/2019] [Accepted: 10/18/2019] [Indexed: 01/09/2023] Open
Abstract
Neuroblastoma (NB) represents one of the most frequent pediatric solid tumors. Macrophage migration inhibitory factor (MIF) is a cytokine exerting multiple biological functions. More recently, a second member of the MIF family of cytokine has been identified, the D-dopachrome tautomerase (DDT), that exerts several overlapping functions with MIF. Growing evidence suggests a key role for MIF and DDT in the development of cancer. The aim of this study is to characterize the prognostic value of MIF and DDT in NB. We show that higher expression levels of MIF and DDT in Stage 4 NB samples are associated with a poorer prognosis, independently of the presence of MYCN amplification. Moreover, higher levels of MIF are mostly enriched by Th1 cells, while lower levels of MIF are associated with an increased proportion of B cells, Cytotoxic T cells, Dendritic cells and Natural Killer T cells. We also show that treatment with the histone deacetylase (HDAC) inhibitor, vorinostat, of the NB cell line, SH-SY5Y, determines a significant reduction in the expression of both MIF and DDT. Finally, MIF and DDT inhibition by short interfering RNA is able to revert vincristine sensitivity in vitro. Overall, our data suggest that MIF exert pro-tumorigenic properties in NB, likely by dampening antigen presentation and cytotoxic immune responses, and we propose the HDAC inhibitors as a potential therapeutic strategy for NB patients.
Collapse
Affiliation(s)
- Eugenio Cavalli
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy.
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy.
| | - Santa Mammana
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy.
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy.
| | - Salvo Danilo Lombardo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy.
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy.
| | - Placido Bramanti
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy.
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy.
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy.
| | | |
Collapse
|
47
|
Lombardo SD, Mazzon E, Basile MS, Campo G, Corsico F, Presti M, Bramanti P, Mangano K, Petralia MC, Nicoletti F, Fagone P. Modulation of Tetraspanin 32 (TSPAN32) Expression in T Cell-Mediated Immune Responses and in Multiple Sclerosis. Int J Mol Sci 2019; 20:ijms20184323. [PMID: 31487788 PMCID: PMC6770290 DOI: 10.3390/ijms20184323] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 08/30/2019] [Indexed: 01/25/2023] Open
Abstract
Tetraspanins are a conserved family of proteins involved in a number of biological processes including, cell–cell interactions, fertility, cancer metastasis and immune responses. It has previously been shown that TSPAN32 knockout mice have normal hemopoiesis and B-cell responses, but hyperproliferative T cells. Here, we show that TSPAN32 is expressed at higher levels in the lymphoid lineage as compared to myeloid cells. In vitro activation of T helper cells via anti-CD3/CD28 is associated with a significant downregulation of TSPAN32. Interestingly, engagement of CD3 is sufficient to modulate TSPAN32 expression, and its effect is potentiated by costimulation with anti-CD28, but not anti-CTLA4, -ICOS nor -PD1. Accordingly, we measured the transcriptomic levels of TSPAN32 in polarized T cells under Th1 and Th2 conditions and TSPAN32 resulted significantly reduced as compared with unstimulated cells. On the other hand, in Treg cells, TSPAN32 underwent minor changes upon activation. The in vitro data were finally translated into the context of multiple sclerosis (MS). Encephalitogenic T cells from Myelin Oligodendrocyte Glycoprotein (MOG)-Induced Experimental Autoimmune Encephalomyelitis (EAE) mice showed significantly lower levels of TSPAN32 and increased levels of CD9, CD53, CD82 and CD151. Similarly, in vitro-activated circulating CD4 T cells from MS patients showed lower levels of TSPAN32 as compared with cells from healthy donors. Overall, these data suggest an immunoregulatory role for TSPAN32 in T helper immune response and may represent a target of future immunoregulatory therapies for T cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Salvo Danilo Lombardo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Giorgia Campo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Federica Corsico
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Mario Presti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Placido Bramanti
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | | | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy.
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| |
Collapse
|
48
|
Lombardo SD, Presti M, Mangano K, Petralia MC, Basile MS, Libra M, Candido S, Fagone P, Mazzon E, Nicoletti F, Bramanti A. Prediction of PD-L1 Expression in Neuroblastoma via Computational Modeling. Brain Sci 2019; 9:E221. [PMID: 31480495 PMCID: PMC6770763 DOI: 10.3390/brainsci9090221] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/26/2019] [Accepted: 08/31/2019] [Indexed: 12/21/2022] Open
Abstract
Immunotherapy is a promising new therapeutic approach for neuroblastoma (NBM): an anti-GD2 vaccine combined with orally administered soluble beta-glucan is undergoing a phase II clinical trial and nivolumab and ipilimumab are being tested in recurrent and refractory tumors. Unfortunately, predictive biomarkers of response to immunotherapy are currently not available for NBM patients. The aim of this study was to create a computational network model simulating the different intracellular pathways involved in NBM, in order to predict how the tumor phenotype may be influenced to increase the sensitivity to anti-programmed cell death-ligand-1 (PD-L1)/programmed cell death-1 (PD-1) immunotherapy. The model runs on COPASI software. In order to determine the influence of intracellular signaling pathways on the expression of PD-L1 in NBM, we first developed an integrated network of protein kinase cascades. Michaelis-Menten kinetics were associated to each reaction in order to tailor the different enzymes kinetics, creating a system of ordinary differential equations (ODEs). The data of this study offers a first tool to be considered in the therapeutic management of the NBM patient undergoing immunotherapeutic treatment.
Collapse
Affiliation(s)
- Salvo Danilo Lombardo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123- Catania, Italy
| | - Mario Presti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123- Catania, Italy
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123- Catania, Italy
| | - Maria Cristina Petralia
- IRCCS (Istituti di Ricovero e Cura a Carattere Scientifico) Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124- Messina, Italy
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123- Catania, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123- Catania, Italy
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123- Catania, Italy
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123- Catania, Italy.
| | - Emanuela Mazzon
- IRCCS (Istituti di Ricovero e Cura a Carattere Scientifico) Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124- Messina, Italy
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123- Catania, Italy
| | - Alessia Bramanti
- IRCCS (Istituti di Ricovero e Cura a Carattere Scientifico) Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124- Messina, Italy
| |
Collapse
|
49
|
Wang L, Zhou Y, Qin Y, Wang Y, Liu B, Fang R, Bai M. Methylophiopogonanone B of Radix Ophiopogonis protects cells from H2O2‑induced apoptosis through the NADPH oxidase pathway in HUVECs. Mol Med Rep 2019; 20:3691-3700. [PMID: 31485606 PMCID: PMC6755187 DOI: 10.3892/mmr.2019.10625] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 07/30/2019] [Indexed: 12/14/2022] Open
Abstract
Methylophiopogonanone B (MO-B), which belongs to a group of homoisoflavonoids, present in Ophiopogon japonicus, has been identified as an active component with antioxidative and anti-tumor properties. The present study investigated whether MO-B may exert protective effects on human umbilical vein endothelial cells (HUVECs) against H2O2-induced injury in vitro, and whether the MO-B effects may be modulated by the NADPH pathway. HUVECs were treated with MO-B in the presence or absence of H2O2. Malondialdehyde (MDA), reactive oxygen species (ROS) levels, and superoxide dismutase (SOD) activity were analyzed to evaluate cell injury and the antioxidative potential of MO-B. The results revealed that MO-B inhibited the production of MDA and ROS, but enhanced SOD activity. Furthermore, MO-B could alleviate H2O2-induced apoptosis in HUVECs, which is consistent with the expression of apoptosis-associated genes and proteins in cells, including Bax/Bcl-2 and caspase-3. To explore the potential mechanism, the present study investigated the effects of MO-B on NADPH-related signaling via the analysis of neutrophil cytochrome b light chain (p22phox) expression, which is the membrane-associated subunit of NADPH oxidase. MO-B could improve the survival of endothelial cells and therefore may be a potential drug in the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Liling Wang
- Zhejiang Academy of Forestry, Zhejiang University of Science and Technology, Hangzhou, Zhejiang 310023, P.R. China
| | - Yifeng Zhou
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, Zhejiang 310023, P.R. China
| | - Yuchuan Qin
- Zhejiang Academy of Forestry, Zhejiang University of Science and Technology, Hangzhou, Zhejiang 310023, P.R. China
| | - Yanbin Wang
- Zhejiang Academy of Forestry, Zhejiang University of Science and Technology, Hangzhou, Zhejiang 310023, P.R. China
| | - Bentong Liu
- Zhejiang Academy of Forestry, Zhejiang University of Science and Technology, Hangzhou, Zhejiang 310023, P.R. China
| | - Ru Fang
- Zhejiang Academy of Forestry, Zhejiang University of Science and Technology, Hangzhou, Zhejiang 310023, P.R. China
| | - Minge Bai
- Zhejiang Academy of Forestry, Zhejiang University of Science and Technology, Hangzhou, Zhejiang 310023, P.R. China
| |
Collapse
|
50
|
Bilsborrow JB, Doherty E, Tilstam PV, Bucala R. Macrophage migration inhibitory factor (MIF) as a therapeutic target for rheumatoid arthritis and systemic lupus erythematosus. Expert Opin Ther Targets 2019; 23:733-744. [PMID: 31414920 DOI: 10.1080/14728222.2019.1656718] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction. Macrophage migration inhibitory factor (MIF) is a pleiotropic inflammatory cytokine with upstream regulatory roles in innate and adaptive immunity and is implicated in the pathogenesis of autoimmune diseases including rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). Several classes of MIF inhibitors such as small molecule inhibitors and peptide inhibitors are in clinical development. Areas covered. The role of MIF in the pathogenesis of RA and SLE is examined; the authors review the structure, physiology and signaling characteristics of MIF and the related cytokine D-DT/MIF-2. The preclinical and clinical trial data for MIF inhibitors are also reviewed; information was retrieved from PubMed and ClinicalTrials.gov using the keywords MIF, D-DT/MIF-2, CD74, CD44, CXCR2, CXCR4, Jab-1, rheumatoid arthritis, systemic lupus erythematosus, MIF inhibitor, small molecule, anti-MIF, anti-CD74, and peptide inhibitor. Expert opinion. Studies in mice and in humans demonstrate the therapeutic potential of MIF inhibition for RA and SLE. MIF- directed approaches could be particularly efficacious in patients with high expression MIF genetic polymorphisms. In patients with RA and SLE and high expression MIF alleles, targeted MIF inhibition could be a precision medicine approach to treatment. Anti-MIF pharmacotherapies could also be steroid-sparing in patients with chronic glucocorticoid dependence or refractory autoimmune disease.
Collapse
Affiliation(s)
- Joshua B Bilsborrow
- Department of Internal Medicine, Yale University School of Medicine , New Haven , CT , USA
| | - Edward Doherty
- Department of Internal Medicine, Yale University School of Medicine , New Haven , CT , USA
| | - Pathricia V Tilstam
- Department of Internal Medicine, Yale University School of Medicine , New Haven , CT , USA
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine , New Haven , CT , USA
| |
Collapse
|