1
|
Chen T, Ma W, Wang X, Ye Q, Hou X, Wang Y, Jiang C, Meng X, Sun Y, Cai J. Insights of immune cell heterogeneity, tumor-initiated subtype transformation, drug resistance, treatment and detecting technologies in glioma microenvironment. J Adv Res 2025; 72:527-554. [PMID: 39097088 DOI: 10.1016/j.jare.2024.07.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/30/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024] Open
Abstract
BACKGROUND With the gradual understanding of glioma development and the immune microenvironment, many immune cells have been discovered. Despite the growing comprehension of immune cell functions and the clinical application of immunotherapy, the precise roles and characteristics of immune cell subtypes, how glioma induces subtype transformation of immune cells and its impact on glioma progression have yet to be understood. AIM OF THE REVIEW In this review, we comprehensively center on the four major immune cells within the glioma microenvironment, particularly neutrophils, macrophages, lymphocytes, myeloid-derived suppressor cells (MDSCs), and other significant immune cells. We discuss (1) immune cell subtype markers, (2) glioma-induced immune cell subtype transformation, (3) the mechanisms of each subtype influencing chemotherapy resistance, (4) therapies targeting immune cells, and (5) immune cell-associated single-cell sequencing. Eventually, we identified the characteristics of immune cell subtypes in glioma, comprehensively summarized the exact mechanism of glioma-induced immune cell subtype transformation, and concluded the progress of single-cell sequencing in exploring immune cell subtypes in glioma. KEY SCIENTIFIC CONCEPTS OF REVIEW In conclusion, we have analyzed the mechanism of chemotherapy resistance detailly, and have discovered prospective immunotherapy targets, excavating the potential of novel immunotherapies approach that synergistically combines radiotherapy, chemotherapy, and surgery, thereby paving the way for improved immunotherapeutic strategies against glioma and enhanced patient outcomes.
Collapse
Affiliation(s)
- Tongzheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenbin Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qile Ye
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xintong Hou
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yiwei Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chuanlu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Six Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangqi Meng
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Ying Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Jinquan Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
2
|
Zhang J, Zhang R, Jin S, Feng X. Curcumin, a plant polyphenol with multiple physiological functions of improving antioxidation, anti-inflammation, immunomodulation and its application in poultry production. J Anim Physiol Anim Nutr (Berl) 2024; 108:1890-1905. [PMID: 39081000 DOI: 10.1111/jpn.14029] [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: 11/25/2023] [Revised: 05/11/2024] [Accepted: 07/18/2024] [Indexed: 11/07/2024]
Abstract
Finding environmentally friendly, effective and residue-free alternatives to antibiotics has become a research priority. This is due to the ban on antibiotics in animal feed. Curcumin is a polyphenol extracted from the rhizome of turmeric that has antioxidant, anti-inflammatory and immunomodulatory properties. Curcumin has been widely demonstrated as a traditional flavoured agent and herbal medicine in the fight against diseases. In recent years, curcumin has been extensively studied in animal production, especially in poultry production. This article reviews the source, structure, metabolism and biological functions of curcumin and focuses on the application of curcumin in poultry production. In terms of production performance, curcumin can improve the growth performance of poultry, increase the egg production rate of laying hens and alleviate the negative effects of heat stress on the production performance of poultry and livestock. In terms of meat quality, curcumin can improve poultry meat quality by regulating lipid metabolism and antioxidant capacity. In terms of health, curcumin can improve immunity. Since mycotoxins have been a major problem in poultry production, this article also reviews the role of curcumin in helping poultry resist toxins. It is hoped that the review in this article can provide a concrete theoretical basis and research ideas for the research and application of curcumin in the field of poultry.
Collapse
Affiliation(s)
- Jingyang Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Ruoshi Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Sanjun Jin
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Xingjun Feng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| |
Collapse
|
3
|
Giordano L, Cacciola R, Barone P, Vecchio V, Nasso ME, Alvaro ME, Gangemi S, Cacciola E, Allegra A. Autoimmune Diseases and Plasma Cells Dyscrasias: Pathogenetic, Molecular and Prognostic Correlations. Diagnostics (Basel) 2024; 14:1135. [PMID: 38893662 PMCID: PMC11171610 DOI: 10.3390/diagnostics14111135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Multiple myeloma and monoclonal gammopathy of undetermined significance are plasma cell dyscrasias characterized by monoclonal proliferation of pathological plasma cells with uncontrolled production of immunoglobulins. Autoimmune pathologies are conditions in which T and B lymphocytes develop a tendency to activate towards self-antigens in the absence of exogenous triggers. The aim of our review is to show the possible correlations between the two pathological aspects. Molecular studies have shown how different cytokines that either cause inflammation or control the immune system play a part in the growth of immunotolerance conditions that make it easier for the development of neoplastic malignancies. Uncontrolled immune activation resulting in chronic inflammation is also known to be at the basis of the evolution toward neoplastic pathologies, as well as multiple myeloma. Another point is the impact that myeloma-specific therapies have on the course of concomitant autoimmune diseases. Indeed, cases have been observed of patients suffering from multiple myeloma treated with daratumumab and bortezomib who also benefited from their autoimmune condition or patients under treatment with immunomodulators in which there has been an arising or worsening of autoimmunity conditions. The role of bone marrow transplantation in the course of concomitant autoimmune diseases remains under analysis.
Collapse
Affiliation(s)
- Laura Giordano
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (L.G.); (P.B.); (V.V.); (M.E.N.); (M.E.A.)
| | - Rossella Cacciola
- Hemostasis/Hematology Unit, Department of Experimental and Clinical Medicine, University of Catania, 95123 Catania, Italy;
| | - Paola Barone
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (L.G.); (P.B.); (V.V.); (M.E.N.); (M.E.A.)
| | - Veronica Vecchio
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (L.G.); (P.B.); (V.V.); (M.E.N.); (M.E.A.)
| | - Maria Elisa Nasso
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (L.G.); (P.B.); (V.V.); (M.E.N.); (M.E.A.)
| | - Maria Eugenia Alvaro
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (L.G.); (P.B.); (V.V.); (M.E.N.); (M.E.A.)
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department and Experimental Medicine, University of Messina, 98125 Messina, Italy;
| | - Emma Cacciola
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy;
| | - Alessandro Allegra
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (L.G.); (P.B.); (V.V.); (M.E.N.); (M.E.A.)
| |
Collapse
|
4
|
Nie SC, Jing YH, Lu L, Ren SS, Ji G, Xu HC. Mechanisms of myeloid-derived suppressor cell-mediated immunosuppression in colorectal cancer and related therapies. World J Gastrointest Oncol 2024; 16:1690-1704. [PMID: 38764816 PMCID: PMC11099432 DOI: 10.4251/wjgo.v16.i5.1690] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/30/2024] [Accepted: 03/11/2024] [Indexed: 05/09/2024] Open
Abstract
Severe immunosuppression is a hallmark of colorectal cancer (CRC). Myeloid-derived suppressor cells (MDSCs), one of the most abundant components of the tumor stroma, play an important role in the invasion, metastasis, and immune escape of CRC. MDSCs create an immunosuppressive microenvironment by inhibiting the proliferation and activation of immunoreactive cells, including T and natural killer cells, as well as by inducing the proliferation of immunosuppressive cells, such as regulatory T cells and tumor-associated macrophages, which, in turn, promote the growth of cancer cells. Thus, MDSCs are key contributors to the emergence of an immunosuppressive microenvironment in CRC and play an important role in the breakdown of antitumor immunity. In this narrative review, we explore the mechanisms through which MDSCs contribute to the immunosuppressive microenvironment, the current therapeutic approaches and technologies targeting MDSCs, and the therapeutic potential of modulating MDSCs in CRC treatment. This study provides ideas and methods to enhance survival rates in patients with CRC.
Collapse
Affiliation(s)
- Shu-Chang Nie
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yan-Hua Jing
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Lu Lu
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, Shanghai 200032, China
| | - Si-Si Ren
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, Shanghai 200032, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine), Shanghai 200032, China
| | - Han-Chen Xu
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, Shanghai 200032, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine), Shanghai 200032, China
| |
Collapse
|
5
|
Kitamura H. Flow cytometric detection of CD11b + Gr-1 + cells in nontumor-bearing mice: A propolis-elicited model. Methods Cell Biol 2023; 184:17-32. [PMID: 38555156 DOI: 10.1016/bs.mcb.2023.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogenous myeloid lineage population whose conventional surface phenotype is CD11b+ Gr-1+. Due to their rarity and fragility, analyses using primary isolated MDSCs are extremely difficult. However, counting CD11b+ Gr-1+ cells in associated tissues such as tumors and inflammatory lesions provides critical information regarding MDSC involvement in immune disorders in the tissues. Specific MDSC markers have not been identified, limiting our ability to apply histochemical approaches during MDSCs research. However, profiling surface antigens using multi-colorimetric flow cytometry enables us to easily monitor the abundance of MDSCs in vivo. Monitoring of mouse MDSCs and their subpopulations using flow cytometry is well established. In this article, I exemplify a conventional method of monitoring CD11b+ Gr-1+ cells in mouse adipose tissue after administration of Brazilian propolis ethanol extract, which is a strong inducer of MDSCs.
Collapse
Affiliation(s)
- Hiroshi Kitamura
- Laboratory of Disease Models, College of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan.
| |
Collapse
|
6
|
Allegra A, Murdaca G, Mirabile G, Gangemi S. Redox Signaling Modulates Activity of Immune Checkpoint Inhibitors in Cancer Patients. Biomedicines 2023; 11:1325. [PMID: 37238995 PMCID: PMC10215686 DOI: 10.3390/biomedicines11051325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 04/23/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Although immunotherapy is already a staple of cancer care, many patients may not benefit from these cutting-edge treatments. A crucial field of research now focuses on figuring out how to improve treatment efficacy and assess the resistance mechanisms underlying this uneven response. For a good response, immune-based treatments, in particular immune checkpoint inhibitors, rely on a strong infiltration of T cells into the tumour microenvironment. The severe metabolic environment that immune cells must endure can drastically reduce effector activity. These immune dysregulation-related tumour-mediated perturbations include oxidative stress, which can encourage lipid peroxidation, ER stress, and T regulatory cells dysfunction. In this review, we have made an effort to characterize the status of immunological checkpoints, the degree of oxidative stress, and the part that latter plays in determining the therapeutic impact of immunological check point inhibitors in different neoplastic diseases. In the second section of the review, we will make an effort to assess new therapeutic possibilities that, by affecting redox signalling, may modify the effectiveness of immunological treatment.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Giuseppe Murdaca
- Department of Internal Medicine, Ospedale Policlinico San Martino IRCCS, University of Genova, Viale Benedetto XV, n. 6, 16132 Genova, Italy
| | - Giuseppe Mirabile
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
| |
Collapse
|
7
|
Allegra A, Mirabile G, Ettari R, Pioggia G, Gangemi S. The Impact of Curcumin on Immune Response: An Immunomodulatory Strategy to Treat Sepsis. Int J Mol Sci 2022; 23:ijms232314710. [PMID: 36499036 PMCID: PMC9738113 DOI: 10.3390/ijms232314710] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/12/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022] Open
Abstract
Primary and secondary immunodeficiencies cause an alteration in the immune response which can increase the rate of infectious diseases and worsened prognoses. They can also alter the immune response, thus, making the infection even worse. Curcumin is the most biologically active component of the turmeric root and appears to be an antimicrobial agent. Curcumin cooperates with various cells such as macrophages, dendritic cells, B, T, and natural killer cells to modify the body's defence capacity. Curcumin also inhibits inflammatory responses by suppressing different metabolic pathways, reduces the production of inflammatory cytokines, and increases the expression of anti-inflammatory cytokines. Curcumin may also affect oxidative stress and the non-coding genetic material. This review analyses the relationships between immunodeficiency and the onset of infectious diseases and discusses the effects of curcumin and its derivatives on the immune response. In addition, we analyse some of the preclinical and clinical studies that support its possible use in prophylaxis or in the treatment of infectious diseases. Lastly, we examine how nanotechnologies can enhance the clinical use of curcumin.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy
- Correspondence:
| | - Giuseppe Mirabile
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy
| | - Roberta Ettari
- Department of Chemical, Biological, Pharmaceutical and Environmental Chemistry, University of Messina, 98100 Messina, Italy
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 98164 Messina, Italy
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| |
Collapse
|
8
|
Bizymi N, Matthaiou AM, Matheakakis A, Voulgari I, Aresti N, Zavitsanou K, Karasachinidis A, Mavroudi I, Pontikoglou C, Papadaki HA. New Perspectives on Myeloid-Derived Suppressor Cells and Their Emerging Role in Haematology. J Clin Med 2022; 11:jcm11185326. [PMID: 36142973 PMCID: PMC9504532 DOI: 10.3390/jcm11185326] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 12/03/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are immature cells of myeloid origin that have gained researchers’ attention, as they constitute promising biomarkers and targets for novel therapeutic strategies (i.e., blockage of development, differentiation, depletion, and deactivation) in several conditions, including neoplastic, autoimmune, infective, and inflammatory diseases, as well as pregnancy, obesity, and graft rejection. They are characterised in humans by the typical immunophenotype of CD11b+CD33+HLA-DR–/low and immune-modulating properties leading to decreased T-cell proliferation, induction of T-regulatory cells (T-regs), hindering of natural killer (NK) cell functionality, and macrophage M2-polarisation. The research in the field is challenging, as there are still difficulties in defining cell-surface markers and gating strategies that uniquely identify the different populations of MDSCs, and the currently available functional assays are highly demanding. There is evidence that MDSCs display altered frequency and/or functionality and could be targeted in immune-mediated and malignant haematologic diseases, although there is a large variability of techniques and results between different laboratories. This review presents the current literature concerning MDSCs in a clinical point of view in an attempt to trigger future investigation by serving as a guide to the clinical haematologist in order to apply them in the context of precision medicine as well as the researcher in the field of experimental haematology.
Collapse
Affiliation(s)
- Nikoleta Bizymi
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
- Laboratory of Molecular and Cellular Pneumonology, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Andreas M. Matthaiou
- Laboratory of Molecular and Cellular Pneumonology, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
- Respiratory Physiology Laboratory, Medical School, University of Cyprus, 2029 Nicosia, Cyprus
| | - Angelos Matheakakis
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Ioanna Voulgari
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Nikoletta Aresti
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Konstantina Zavitsanou
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Anastasios Karasachinidis
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Irene Mavroudi
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Charalampos Pontikoglou
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Helen A. Papadaki
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
- Correspondence: ; Tel.: +30-2810394637
| |
Collapse
|
9
|
Allegra A, Casciaro M, Lo Presti E, Musolino C, Gangemi S. Harnessing Unconventional T Cells and Innate Lymphoid Cells to Prevent and Treat Hematological Malignancies: Prospects for New Immunotherapy. Biomolecules 2022; 12:biom12060754. [PMID: 35740879 PMCID: PMC9221132 DOI: 10.3390/biom12060754] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/21/2022] [Accepted: 05/26/2022] [Indexed: 11/18/2022] Open
Abstract
Unconventional T cells and innate lymphoid cells (ILCs) make up a heterogeneous set of cells that characteristically show prompt responses toward specific antigens. Unconventional T cells recognize non-peptide antigens, which are bound and presented by diverse non-polymorphic antigen-presenting molecules and comprise γδ T cells, MR1-restricted mucosal-associated invariant T cells (MAITs), and natural killer T cells (NKTs). On the other hand, ILCs lack antigen-specific receptors and act as the innate counterpart to the T lymphocytes found in the adaptive immune response. The alteration of unconventional T cells and ILCs in frequency and functionality is correlated with the onset of several autoimmune diseases, allergy, inflammation, and tumor. However, depending on the physio-pathological framework, unconventional T cells may exhibit either protective or pathogenic activity in a range of neoplastic diseases. Nonetheless, experimental models and clinical studies have displayed that some unconventional T cells are potential therapeutic targets, as well as prognostic and diagnostic markers. In fact, cell-mediated immune response in tumors has become the focus in immunotherapy against neoplastic disease. This review concentrates on the present knowledge concerning the function of unconventional T cell sets in the antitumor immune response in hematological malignancies, such as acute and chronic leukemia, multiple myeloma, and lymphoproliferative disorders. Moreover, we discuss the possibility that modulating the activity of unconventional T cells could be useful in the treatment of hematological neoplasms, in the prevention of specific conditions (such as graft versus host disease), and in the formulation of an effective anticancer vaccine therapy. The exact knowledge of the role of these cells could represent the prerequisite for the creation of a new form of immunotherapy for hematological neoplasms.
Collapse
Affiliation(s)
- Alessandro Allegra
- Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, Division of Hematology, University of Messina, 98125 Messina, Italy; (A.A.); (C.M.)
| | - Marco Casciaro
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy;
- Correspondence: ; Tel.: +39-090-221-2013
| | - Elena Lo Presti
- National Research Council (CNR)—Institute for Biomedical Research and Innovation (IRIB), 90146 Palermo, Italy;
| | - Caterina Musolino
- Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, Division of Hematology, University of Messina, 98125 Messina, Italy; (A.A.); (C.M.)
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy;
| |
Collapse
|
10
|
Allegra A, Di Gioacchino M, Tonacci A, Petrarca C, Musolino C, Gangemi S. Multiple Myeloma Cell-Derived Exosomes: Implications on Tumorigenesis, Diagnosis, Prognosis and Therapeutic Strategies. Cells 2021; 10:2865. [PMID: 34831088 PMCID: PMC8616233 DOI: 10.3390/cells10112865] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/16/2022] Open
Abstract
Multiple myeloma (MM) is a hematological disease that is still not curable. The bone marrow milieu, with cellular and non-cellular elements, participate in the creation of a pro-tumoral environment enhancing growth and survival of MM plasma cells. Exosomes are vesicles oscillating in dimension between 50 nm and 100 nm in size that can be released by various cells and contribute to the pathogenesis and progression of MM. Exosomes enclose proteins, cytokines, lipids, microRNAs, long noncoding RNAs, and circular RNAs able to regulate interactions between MM plasma cells and adjacent cells. Through exosomes, mesenchymal stem cells confer chemoresistance to MM cells, while myeloma cells promote angiogenesis, influence immune response, cause bone lesions, and have an impact on the outcome of MM patients. In this review, we analyze the role played by exosomes in the progression of monoclonal gammopathies and the effects on the proliferation of neoplastic plasma cells, and discuss the possible employment of exosomes as potential targets for the treatment of MM patients.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Mario Di Gioacchino
- Center for Advanced Studies and Technology, G. D’Annunzio University, 66100 Chieti, Italy;
- Institute for Clinical Immunotherapy and Advanced Biological Treatments, 65100 Pescara, Italy
| | - Alessandro Tonacci
- National Research Council of Italy (IFC-CNR), Clinical Physiology Institute, 56124 Pisa, Italy;
| | - Claudia Petrarca
- Center for Advanced Studies and Technology, G. D’Annunzio University, 66100 Chieti, Italy;
- Institute for Clinical Immunotherapy and Advanced Biological Treatments, 65100 Pescara, Italy
- National Research Council of Italy (IFC-CNR), Clinical Physiology Institute, 56124 Pisa, Italy;
- Department of Medicine and Science of Ageing, G. D’Annunzio University, 66100 Chieti, Italy
| | - Caterina Musolino
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, Unit and School of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy;
| |
Collapse
|
11
|
Nanomedicine for Immunotherapy Targeting Hematological Malignancies: Current Approaches and Perspective. NANOMATERIALS 2021; 11:nano11112792. [PMID: 34835555 PMCID: PMC8619332 DOI: 10.3390/nano11112792] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/04/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022]
Abstract
Conventional chemotherapy has partial therapeutic effects against hematological malignancies and is correlated with serious side effects and great risk of relapse. Recently, immunotherapeutic drugs have provided encouraging results in the treatment of hematological malignancies. Several immunotherapeutic antibodies and cell therapeutics are in dynamic development such as immune checkpoint blockades and CAR-T treatment. However, numerous problems restrain the therapeutic effectiveness of tumor immunotherapy as an insufficient anti-tumor immune response, the interference of an immune-suppressive bone marrow, or tumoral milieu with the discharge of immunosuppressive components, access of myeloid-derived suppressor cells, monocyte intrusion, macrophage modifications, all factors facilitating the tumor to escape the anti-cancer immune response, finally reducing the efficiency of the immunotherapy. Nanotechnology can be employed to overcome each of these aspects, therefore having the possibility to successfully produce anti-cancer immune responses. Here, we review recent findings on the use of biomaterial-based nanoparticles in hematological malignancies immunotherapy. In the future, a deeper understanding of tumor immunology and of the implications of nanomedicine will allow nanoparticles to revolutionize tumor immunotherapy, and nanomedicine approaches will reveal their great potential for clinical translation.
Collapse
|
12
|
Liu T, Yang F, Xie J, Chen J, Gao W, Bai X, Li Z. All-trans-retinoic acid restores CD4+ T cell response after sepsis by inhibiting the expansion and activation of myeloid-derived suppressor cells. Mol Immunol 2021; 136:8-15. [PMID: 34051632 DOI: 10.1016/j.molimm.2021.04.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/28/2021] [Accepted: 04/26/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Patients are susceptible to immunosuppression in late-stage of sepsis, in which myeloid-derived suppressor cells (MDSCs) is an important contributor. This study aims to investigate whether all-trans-retinoic acid (ATRA), which has been proved to inhibit MDSCs generation in cancer, will ameliorate sepsis-induced immuno-suppression through modulating MDSCs. METHODS A clinically relevant "two-hit'' model of sepsis, the cecal ligation and puncture (CLP) model and secondary pneumonia model, were established in mice. The effects of ATRA on the mortality, the bacterial burden, the expansion and activity of CLP-induced MDSCs, as well as the function of CD4+ T cells were evaluated. RESULTS In CLP model, ATRA was found to reduce frequency of MDSCs in spleen of mice and inhibit activity of MDSCs by regulating the generation and activity of arginase-1 and iNOS, and the secretion of immune-supressive cytokines. ATRA administration eventually reduced mortality of secondary infection by Legionella pneumophila in CLP-surviving mice, which might be associated with the restoration of CD4+ T cells proliferating and secreting activity. CONCLUSION ATRA can restore CD4+ T cells dysfunction in sepsis by modulating the expansion and function of MDSCs and therefore provides a potential therapy that targets the immunosuppressive state of sepsis.
Collapse
Affiliation(s)
- Tao Liu
- Department of Trauma Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, China
| | - Fan Yang
- Department of Trauma Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, China
| | - Jie Xie
- Department of Trauma Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, China
| | - Jiajun Chen
- Department of Trauma Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, China
| | - Wei Gao
- Department of Trauma Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, China
| | - Xiangjun Bai
- Department of Trauma Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, China
| | - Zhanfei Li
- Department of Trauma Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, China.
| |
Collapse
|
13
|
Innao V, Allegra A, Ginaldi L, Pioggia G, De Martinis M, Musolino C, Gangemi S. Reviewing the Significance of Vitamin D Substitution in Monoclonal Gammopathies. Int J Mol Sci 2021; 22:4922. [PMID: 34066482 PMCID: PMC8124934 DOI: 10.3390/ijms22094922] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 11/25/2022] Open
Abstract
Vitamin D is a steroid hormone that is essential for bone mineral metabolism and it has several other effects in the body, including anti-cancer actions. Vitamin D causes a reduction in cell growth by interrupting the cell cycle. Moreover, the active form of vitamin D, i.e., 1,25-dihydroxyvitamin D, exerts various effects via its interaction with the vitamin D receptor on the innate and adaptive immune system, which could be relevant in the onset of tumors. Multiple myeloma is a treatable but incurable malignancy characterized by the growth of clonal plasma cells in protective niches in the bone marrow. In patients affected by multiple myeloma, vitamin D deficiency is commonly correlated with an advanced stage of the disease, greater risk of progression, the development of pathological fractures, and a worse prognosis. Changes in the vitamin D receptor often contribute to the occurrence and progress of deficiencies, which can be overcome by supplementation with vitamin D or analogues. However, in spite of the findings available in the literature, there is no clear standard of care and clinical practice varies. Further research is needed to better understand how vitamin D influences outcomes in patients with monoclonal gammopathies.
Collapse
Affiliation(s)
- Vanessa Innao
- Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, Division of Haematology, University of Messina, 98125 Messina, Italy; (V.I.); (C.M.)
| | - Alessandro Allegra
- Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, Division of Haematology, University of Messina, 98125 Messina, Italy; (V.I.); (C.M.)
| | - Lia Ginaldi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (L.G.); (M.D.M.)
- Allergy and Clinical Immunology Unit, Center for the Diagnosis and Treatment of Osteoporosis, AUSL 04 Teramo, 64100 Teramo, Italy
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 98164 Messina, Italy;
| | - Massimo De Martinis
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (L.G.); (M.D.M.)
- Allergy and Clinical Immunology Unit, Center for the Diagnosis and Treatment of Osteoporosis, AUSL 04 Teramo, 64100 Teramo, Italy
| | - Caterina Musolino
- Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, Division of Haematology, University of Messina, 98125 Messina, Italy; (V.I.); (C.M.)
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy;
| |
Collapse
|
14
|
Tian S, Liao L, Zhou Q, Huang X, Zheng P, Guo Y, Deng T, Tian X. Curcumin inhibits the growth of liver cancer by impairing myeloid-derived suppressor cells in murine tumor tissues. Oncol Lett 2021; 21:286. [PMID: 33732362 PMCID: PMC7905673 DOI: 10.3892/ol.2021.12547] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 01/14/2021] [Indexed: 12/13/2022] Open
Abstract
Curcumin, one of the active ingredients of Curcuma longa (Jianghuang), has been reported to exert multiple bioactivities, including pro-apoptotic and anti-inflammatory activities. In recent years, curcumin has been extensively studied, and it has been revealed that curcumin inhibits the growth of numerous types of cancer. However, to the best of our knowledge, the inhibitory effects of curcumin on the activation or expansion of myeloid-derived suppressor cells (MDSCs) in liver cancer and the underlying mechanism have not yet been determined. Therefore, the present study aimed to investigate the inhibitory effect of curcumin on MDSC activity and the associated anti-neoplastic mechanism in a HepG2 ×enograft mouse model. The effect of curcumin on the viability of Huh-7, MHCC-97H and HepG2 cells in vitro was analyzed using a Cell Counting Kit-8 assay. The effects of curcumin on tumor growth, numbers of MDSCs, expression levels of proteins involved in the toll-like receptor 4 (TLR4)/NF-κB signaling pathway, levels of related inflammatory factors and angiogenesis were determined in HepG2 ×enograft model mice, which were given different doses of curcumin via intragastrical administration. The results of the present study revealed that curcumin inhibited the viability of Huh-7, MHCC-97H and HepG2 cells and the growth of HepG2 ×enograft tumors in mice. Flow cytometric analysis indicated that curcumin reduced the number of MDSCs in mouse xenograft tumors. In addition, the results demonstrated that curcumin inhibited the TLR4/NF-κB signaling pathway and the expression of inflammatory factors, including IL-6, IL-1β, prostaglandin E2 and cyclooxygenase-2, in mouse xenograft tumors. Furthermore, curcumin suppressed the secretion of granulocyte-macrophage colony-stimulating factor (GM-CSF) and granulocyte-colony stimulating factor (G-CSF), which are essential factors for MDSCs modulation, in tumor tissues. Additionally, curcumin was revealed to inhibit angiogenesis, which was demonstrated by the downregulation of the expression levels of vascular endothelial growth factor, CD31 and α-smooth muscle actin in western blotting, immunohistochemistry and immunofluorescence experiments. In conclusion, the findings of the present study identified a novel mechanism via which curcumin may suppress the growth of liver cancer by reducing the numbers of MDSCs and subsequently disrupting the process of angiogenesis. These conclusions were supported by the observed inactivation of the TLR4/NF-κB signaling pathway-mediated inflammatory response and the downregulation of GM-CSF and G-CSF secretion in xenograft tissues.
Collapse
Affiliation(s)
- Sha Tian
- Department of Internal Medicine, College of Integrated Chinese and Western Medicine of Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Liu Liao
- Department of Internal Medicine, College of Integrated Chinese and Western Medicine of Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Qing Zhou
- Department of Andrology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Xiaodi Huang
- Hunan Key Laboratory of Traditional Chinese Medicine Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Piao Zheng
- Department of Internal Medicine, College of Integrated Chinese and Western Medicine of Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Yinmei Guo
- Department of Internal Medicine, College of Integrated Chinese and Western Medicine of Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Tianhao Deng
- Department of Oncology, The Affiliated Hospital of Hunan Institute of Chinese Medicine, Changsha, Hunan 410006, P.R. China
| | - Xuefei Tian
- Department of Internal Medicine, College of Integrated Chinese and Western Medicine of Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| |
Collapse
|
15
|
Liao TY, Liaw CC. Retrospective Analysis of Mortality Cases in Advanced and Metastatic Solid Tumors With Concurrent Prerenal Azotemia. In Vivo 2021; 34:1515-1519. [PMID: 32354956 DOI: 10.21873/invivo.11939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM A retrospective study of cases with metastatic or advanced solid tumors complicated with AKI (acute kidney injury) with prerenal azotemia. PATIENTS AND METHODS Criteria included: (1) advanced or metastatic solid tumors that led to mortality; (2) prerenal azotemia identified upon renal function evaluation and (3) BUN to Cr ratio (BCR)≥15. We also compared the outcomes of patients with BCR>20 with those of patients with BCR=15-20. RESULTS A total of 218 patients with solid tumors were enrolled. One hundred and forty (64%) and 78 (36%) patients had BCR>20 and 15-20, respectively. Before AKI occurrence, 136 (62%) had thromboembolic complications and 96 (44%) paraneoplastic syndromes. Median survival time was 1 week in all patients. Median survival time was statistically different between the groups with BCR15-20 and BCR>20 (p<0.005, log-rank test). CONCLUSION Cancer patients with concurrent AKI and prerenal azotemia carry a very poor prognosis.
Collapse
Affiliation(s)
- Tzu-Yao Liao
- Division of Hemato-Oncology, Department of Internal Medicine, Chang-Gung Memorial Hospital and Chang-Gung University College of Medicine, Taoyuan, Taiwan, R.O.C
| | - Chuang-Chi Liaw
- Division of Hemato-Oncology, Department of Internal Medicine, Chang-Gung Memorial Hospital and Chang-Gung University College of Medicine, Taoyuan, Taiwan, R.O.C.
| |
Collapse
|
16
|
Early Posttransplant Mobilization of Monocytic Myeloid-derived Suppressor Cell Correlates With Increase in Soluble Immunosuppressive Factors and Predicts Cancer in Kidney Recipients. Transplantation 2021; 104:2599-2608. [PMID: 32068661 DOI: 10.1097/tp.0000000000003179] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Myeloid-derived suppressor cells (MDSCs) increase in patients with cancer and are associated with poor prognosis; however, their role in transplantation is not yet understood. Here we aimed to study the MDSC effects on the evolution of kidney transplant recipients (KTRs). METHODS A cohort of 229 KTRs was prospectively analyzed. Two myeloid cells subsets. CD11bCD33CD14CD15HLA-DR (monocytic MDSC [M-MDSC]) and CD11bCD33CD14CD15HLA-DR (monocytes), were defined by flow cytometry. The suppressive capacity of myeloid cells was tested in cocultures with autologous lymphocytes. Suppressive soluble factors, cytokines, anti-HLA antibodies, and total antioxidant capacity were quantified in plasma. RESULTS Pretransplant, M-MDSC, and monocytes were similar in KTRs and healthy volunteers. M-MDSCs increased immediately posttransplantation and suppressed CD4 and CD8 T cells proliferation. M-MDSCs remained high for 1 y posttransplantation. Higher M-MDSC counts at day 14 posttransplant were observed in patients who subsequently developed cancer, and KTRs with higher M-MDSC at day 14 had significantly lower malignancy-free survival. Day 14 M-MDSC >179.2 per microliter conferred 6.98 times (95% confidence interval, 1.28-37.69) more risk to develop cancer, independently from age, gender, and immunosuppression. Early posttransplant M-MDSCs were lower in patients with enhanced alloimmune response as represented by anti-HLA sensitization. M-MDSC counts correlated with higher circulatory suppressive factors arginase-1 and interleukin-10, and lower total antioxidant capacity. CONCLUSIONS Early posttransplant mobilization of M-MDSCs predicts cancer and adds risk as an independent factor. M-MDSC may favor an immunosuppressive environment that promotes tumoral development.
Collapse
|
17
|
Phan T, Nguyen VH, Buettner R, Morales C, Yang L, Wong P, Tsai W, Salazar MD, Gil Z, Diamond DJ, Rabinowitz JD, Rosen S, Melstrom LG. Inhibition of de novo pyrimidine synthesis augments Gemcitabine induced growth inhibition in an immunocompetent model of pancreatic cancer. Int J Biol Sci 2021; 17:2240-2251. [PMID: 34239352 PMCID: PMC8241727 DOI: 10.7150/ijbs.60473] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/18/2021] [Indexed: 12/13/2022] Open
Abstract
Leflunomide (Lef) is an agent used in autoimmune disorders that interferes with DNA synthesis. De Novo pyrimidine synthesis is a mechanism of Gemcitabine (Gem) resistance in pancreatic cancer. This study aims to assess the efficacy and changes in the tumor microenvironment of Lef monotherapy and in combination with Gem, in a syngeneic mouse model of pancreatic cancer. Methods: MTS proliferation assays were conducted to assess growth inhibition by Gem (0-20 nM), Lef (0-40 uM) and Gem+Lef in KPC (KrasLSL.G12D/+;p53R172H/+; PdxCretg/+) cells in vitro. An in vivo heterotopic KPC model was used and cohorts were treated with: PBS (control), Gem (75 mg/kg/q3d), Lef (40 mg/kg/d), or Gem+Lef. At d28 post-treatment, tumor burden, proliferation index (Ki67), and vascularity (CD31) were measured. Changes in the frequency of peripheral and intratumoral immune cell subsets were evaluated via FACS. Liquid chromatography-mass spectrometry was used for metabolomics profiling. Results: Lef inhibits KPC cell growth and synergizes with Gem in vitro (P<0.05; Combination Index 0.44 (<1 indicates synergy). In vivo, Lef alone and in combination with Gem delays KPC tumor progression (P<0.001). CTLA-4+T cells are also significantly decreased in tumors treated with Lef, Gem or in combination (Gem+Lef) compared to controls (P<0.05). Combination therapy also decreased the Ki67 and vascularity (P<0.01). Leflunomide inhibits de novo pyrimidine synthesis both in vitro (p<0.0001) and in vivo (p<0.05). Conclusions: In this study, we demonstrated that Gem+Lef inhibits pancreatic cancer growth, decrease T cell exhaustion, vascularity and as proof of principle inhibits de novo pyrimidine synthesis. Further characterization of changes in adaptive immunity are necessary to characterize the mechanism of tumor growth inhibition and facilitate translation to a clinical trial.
Collapse
Affiliation(s)
- Thuy Phan
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010
| | - Vu H. Nguyen
- Department of Hematology, City of Hope National Medical Center, Duarte, CA 91010
| | - Ralf Buettner
- Department of Hematology, City of Hope National Medical Center, Duarte, CA 91010
| | - Corey Morales
- Department of Hematology, City of Hope National Medical Center, Duarte, CA 91010
| | - Lifeng Yang
- Lewis Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University, Princeton, NJ 08544
| | - Paul Wong
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010
| | - Weiman Tsai
- Department of Hematology, City of Hope National Medical Center, Duarte, CA 91010
| | | | - Ziv Gil
- Rambam Medical Center, Israel
| | - Don J Diamond
- Department of Hematology, City of Hope National Medical Center, Duarte, CA 91010
| | - Joshua D. Rabinowitz
- Lewis Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University, Princeton, NJ 08544
| | - Steven Rosen
- Department of Hematology, City of Hope National Medical Center, Duarte, CA 91010
| | - Laleh G. Melstrom
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010
- ✉ Corresponding author: Laleh Melstrom MD, City of Hope National Medical Center, Department of Surgery and Immuno-oncology, 1500 E Duarte Road, Duarte, CA 91010. E-mail: ; Phone: 626 218 0282; Fax: 626 218 1113
| |
Collapse
|
18
|
Sasidharan Nair V, Saleh R, Toor SM, Alajez NM, Elkord E. Transcriptomic Analyses of Myeloid-Derived Suppressor Cell Subsets in the Circulation of Colorectal Cancer Patients. Front Oncol 2020; 10:1530. [PMID: 32984004 PMCID: PMC7492613 DOI: 10.3389/fonc.2020.01530] [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: 06/10/2020] [Accepted: 07/16/2020] [Indexed: 12/24/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) promote tumor immune evasion and favor tumorigenesis by activating various tumor-promoting downstream signals. MDSC expansion is evident in the circulation and tumor microenvironment of many solid tumors including colorectal cancer (CRC). We have recently reported the transcriptomic profiles of tumor-infiltrating MDSCs in CRC patients and uncovered pathways, which could potentially assist tumor progression. In this study, we sorted different subsets of circulating MDSCs in CRC patients and investigated their transcriptomic profiles in order to disclose pathways, which could potentially contribute to disease progression. The sorted subsets included polymorphonuclear/granulocytic MDSCs (PMN-MDSCs), immature MDSCs (I-MDSCs), and monocytic MDSCs (M-MDSCs). Our functional annotation analyses revealed that multiple pathways including DNA damage-, chemotaxis-, apoptosis-, mitogen-activated protein kinase-, transforming growth factor β-, and myeloid differentiation–related transcripts were higher in PMN-MDSCs, compared with monocytic antigen-presenting cells (APCs) or I-MDSCs. Furthermore, genes related to Janus kinase (JAK)–signal transducer and activator of transcription (STAT) were also elevated in PMN-MDSCs. These data suggest that upregulation of JAK-STAT pathway could trigger multiple downstream targets in PMN-MDSCs, which favor tumor progression. Additionally, we found that pathways including phosphatidyl inositol 3-kinase (PI3K), interleukin 6, and TGF-β in M-MDSCs and cell cycle–related pathways in I-MDSCs were upregulated, compared with monocytic APCs. Moreover, acetylation-related genes were upregulated in both PMN-MDSCs and M-MDSCs. This latter finding implicates that epigenetic modifications could also play a role in the regulation of multiple tumor-promoting genes in PMN-MDSCs and M-MDSCs. Taken together, this study reveals various signaling pathways, which regulate the function of MDSC subsets in the circulation of CRC patients. However, functional studies are warranted to support these findings.
Collapse
Affiliation(s)
- Varun Sasidharan Nair
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Reem Saleh
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Salman M Toor
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Nehad M Alajez
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Eyad Elkord
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| |
Collapse
|
19
|
Allegra A, Tonacci A, Pioggia G, Musolino C, Gangemi S. Anticancer Activity of Rosmarinus officinalis L.: Mechanisms of Action and Therapeutic Potentials. Nutrients 2020; 12:E1739. [PMID: 32532056 PMCID: PMC7352773 DOI: 10.3390/nu12061739] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 12/16/2022] Open
Abstract
Alternative treatments for neoplastic diseases with new drugs are necessary because the clinical effectiveness of chemotherapy is often reduced by collateral effects. Several natural substances of plant origin have been demonstrated to be successful in the prevention and treatment of numerous tumors. Rosmarinus officinalis L. is a herb that is cultivated in diverse areas of the world. There is increasing attention being directed towards the pharmaceutical capacities of rosemary, utilized for its anti-inflammatory, anti-infective or anticancer action. The antitumor effect of rosemary has been related to diverse mechanisms, such as the antioxidant effect, antiangiogenic properties, epigenetic actions, regulation of the immune response and anti-inflammatory response, modification of specific metabolic pathways, and increased expression of onco-suppressor genes. In this review, we aim to report the results of preclinical studies dealing with the anticancer effects of rosemary, the molecular mechanisms related to these actions, and the interactions between rosemary and anticancer drugs. The prospect of utilizing rosemary as an agent in the treatment of different neoplastic diseases is discussed. However, although the use of rosemary in the therapy of neoplasms constitutes a fascinating field of study, large and controlled studies must be conducted to definitively clarify the real impact of this substance in clinical practice.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Alessandro Tonacci
- Clinical Physiology Institute, National Research Council of Italy (IFC-CNR), 56124 Pisa, Italy;
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 98164 Messina, Italy;
| | - Caterina Musolino
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
| |
Collapse
|
20
|
Xie Y, Zhang Y, Wei X, Zhou C, Huang Y, Zhu X, Chen Y, Wen H, Huang X, Lin J, Wang Z, Ren Y, Fan B, Deng X, Tan W, Wang C. Jianpi Huayu Decoction Attenuates the Immunosuppressive Status of H 22 Hepatocellular Carcinoma-Bearing Mice: By Targeting Myeloid-Derived Suppressor Cells. Front Pharmacol 2020; 11:16. [PMID: 32140106 PMCID: PMC7042893 DOI: 10.3389/fphar.2020.00016] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 01/07/2020] [Indexed: 12/12/2022] Open
Abstract
Tumor-induced immunosuppressive microenvironment in which myeloid-derived suppressor cells (MDSCs) plays an important role, remains an obstacle for effective oncotherapy currently. Inducing MDSCs into maturation was confirmed as an effective method to reduce the tumor-bearing host's immunosuppression. Traditional Chinese medicines (TCM) possess characteristics of alleviating immunosuppression of cancer patients and low toxicity. Jianpi Huayu Decoction (JHD) was an experienced formula of TCM for oncotherapy based on TCM theory and clinical practice. We previously observed that JHD attenuated the expression of interleukin-10 (IL-10) and transforming growth factor beta (TGF-β) in tumor. IL-10 and TGF-β were found to be cytokines positively related to immunosuppression induced by MDSCs. Here, our study was designed to further investigate the regulation of JHD on the immune system in the H22 liver-cancer mouse model. Mainly, flow cytometry was used to detect the proportion of immune cells, to analyze the apoptosis, differentiation and reactive oxygen species of MDSCs. We found that JHD significantly reduced the destruction of spleen structure, reduced the proportion of regulatory T cells (Treg) and T helper 17 cells (Th17), and increased the proportion of cytotoxic T lymphotes (CTL), Dendritic cells (DC) and CD11b+Gr-1+cells in spleen, but with no significant change of T helper 1 cells (Th1), T helper 2 cells (Th2) and macrophages. In vitro experiments showed that apoptosis of MDSCs was decreased as the time of JHD stimulation increased, which partly explained the increase of CD11b+Gr-1+cells in the spleen. Meanwhile, JHD could promote the differentiation of MDSCs into macrophages and dendritic cells, attenuate expression of ROS in MDSCs and reduce its inhibition on the proliferation of CD4+ T cells, in vitro. Therefore, that the proportion of CD11b+Gr-1+ cells increased in the spleen of tumor-bearing hosts may not be villainy after treatment, when these drugs suppress the immunosuppressive ability of CD11b+Gr-1+ cells and promote it mature to replenish dendritic cell, at the same time. Generally, JHD may be a complementary and alternative drug for attenuating the immunosuppressive status induced by hepatocellular carcinoma, possibly by promoting differentiation and inhibiting the immunosuppressive activity of MDSCs.
Collapse
Affiliation(s)
- Yingjie Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, China
| | - Yuan Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, China
| | - Xiaohan Wei
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Cheng Zhou
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Yajing Huang
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, China.,Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xingwang Zhu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yongxu Chen
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, China
| | - Huihong Wen
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, China
| | - Xuhui Huang
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, China
| | - Juze Lin
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, China
| | - Ziying Wang
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Yan Ren
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, China.,Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Baochao Fan
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, China.,Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xue Deng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Wei Tan
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, China
| | - Changjun Wang
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, China.,School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
21
|
Wang L, Niu Z, Wang X, Li Z, Liu Y, Luo F, Yan X. PHD2 exerts anti-cancer and anti-inflammatory effects in colon cancer xenografts mice via attenuating NF-κB activity. Life Sci 2019; 242:117167. [PMID: 31838134 DOI: 10.1016/j.lfs.2019.117167] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 11/29/2019] [Accepted: 12/09/2019] [Indexed: 02/05/2023]
Abstract
Recent studies suggested that prolyl hydroxylase 2 (PHD2) functions as an important regulator in vascular inflammation and Streptococcus pneumonia infection. However, whether PHD2 contributed to tumor progression prompted by intratumoral inflammation remains elusive. In this study, the effects of PHD2 in colon cancer were evaluated, and the underlying molecular mechanisms were investigated. The results showed that overexpressing PHD2 exerted proliferative and migratory inhibition in colon cancer cells. The expression of cell cycle and epithelial-mesenchymal transition (EMT)-associated proteins were changed: CyclinD1, CDK4, N-cadherin, and Vimentin were down-regulated, while E-cadherin was up-regulated in PHD2-overexpressing colon cancer cells. Moreover, in colon cancer xenograft mice, PHD2 overexpression suppressed tumor growth accompanied by decreased Ki67 expression. Importantly, we further demonstrated that overexpressing PHD2 attenuated inflammation in colon cancer xenograft mice through weakening accumulation of myeloid-derived suppressor cells (MDSCs) and M2-like tumor-associated macrophages (TAMs), as well as secretions of pro-inflammatory cytokines including G-CSF, TNF-α, IL-6, IL-8, IL-1β, and IL-4. Mechanistically, PHD2 overexpression obviously suppressed NF-κB activity through decreasing phosphorylated IκB-α while increasing cytoplasmic NF-κB p65 levels in colon cancer. Our findings support the anti-cancer and anti-inflammatory roles of PHD2 and offer a preclinical proof of tumor progression regulated by cancer cells and inflammation.
Collapse
Affiliation(s)
- Li Wang
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zhendong Niu
- Department of Emergency Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xia Wang
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China; Department of Medical Oncology, Ganzhou City People's Hospital, Ganzhou, Jiangxi, China
| | - Zhixi Li
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yanyang Liu
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Feng Luo
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| | - Xi Yan
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
22
|
The ST2/Interleukin-33 Axis in Hematologic Malignancies: The IL-33 Paradox. Int J Mol Sci 2019; 20:ijms20205226. [PMID: 31652497 PMCID: PMC6834139 DOI: 10.3390/ijms20205226] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 02/07/2023] Open
Abstract
Interleukin (IL)-33 is a chromatin-related nuclear interleukin that is a component of IL-1 family. IL-33 production augments the course of inflammation after cell damage or death. It is discharged into the extracellular space. IL-33 is regarded as an “alarmin” able to stimulate several effectors of the immune system, regulating numerous immune responses comprising cancer immune reactions. IL-33 has been demonstrated to influence tumorigenesis. However, as far as this cytokine is concerned, we are faced with what has sometimes been defined as the IL-33 paradox. Several studies have demonstrated a relevant role of IL-33 to numerous malignancies, where it may have pro- and—less frequently—antitumorigenic actions. In the field of hematological malignancies, the role of IL-33 seems even more complex. Although we can affirm the existence of a negative role of IL-33 in Chronic myelogenos leukemia (CML) and in lymphoproliferative diseases and a positive role in pathologies such as Acute myeloid leukemia (AML), the action of IL-33 seems to be multiple and sometimes contradictory within the same pathology. In the future, we will have to learn to govern the negative aspects of activating the IL-33/ST2 axis and exploit the positive ones.
Collapse
|
23
|
Othman N, Jamal R, Abu N. Cancer-Derived Exosomes as Effectors of Key Inflammation-Related Players. Front Immunol 2019; 10:2103. [PMID: 31555295 PMCID: PMC6737008 DOI: 10.3389/fimmu.2019.02103] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 08/20/2019] [Indexed: 12/11/2022] Open
Abstract
Exosomes, a category of small lipid bilayer extracellular vesicles that are naturally secreted by many cells (both healthy and diseased), carry cargo made up of proteins, lipids, DNAs, and RNAs; all of which are functional when transferred to their recipient cells. Numerous studies have demonstrated the powerful role that exosomes play in the mediation of cell-to-cell communication to induce a pro-tumoral environment to encourage tumor progression and survival. Recently, considerable interest has developed in regard to the role that exosomes play in immunity; with studies demonstrating the ability of exosomes to either metabolically alter immune players such as dendritic cells, T cells, macrophages, and natural killer cells. In this review, we summarize the recent literature on the function of exosomes in regulating a key process that has long been associated with the progression of cancer-inflammation and immunity.
Collapse
Affiliation(s)
- Norahayu Othman
- UKM Medical Centre, UKM Medical Molecular Biology Institute, Kuala Lumpur, Malaysia
| | - Rahman Jamal
- UKM Medical Centre, UKM Medical Molecular Biology Institute, Kuala Lumpur, Malaysia
| | - Nadiah Abu
- UKM Medical Centre, UKM Medical Molecular Biology Institute, Kuala Lumpur, Malaysia
| |
Collapse
|
24
|
Agrati C, Tumino N, Bordoni V, Pinnetti C, Sabatini A, Amendola A, Abbate I, Lorenzini P, Mondi A, Casetti R, Cimini E, Grassi G, Antinori A, Sacchi A. Myeloid Derived Suppressor Cells Expansion Persists After Early ART and May Affect CD4 T Cell Recovery. Front Immunol 2019; 10:1886. [PMID: 31440256 PMCID: PMC6694843 DOI: 10.3389/fimmu.2019.01886] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/25/2019] [Indexed: 12/26/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) are expanded during HIV-1 infection and correlated with disease progression. MDSC expand in the early phase of primary infection depending on TRAIL level. In this study we evaluated the effect of ART on the frequency of MDSC in patients with primary HIV infection (PHI), and their impact on CD4 T cell reconstitution. MDSC frequency was evaluated by flow-cytometry in 60 PHI patients at 12, 24 and 48 weeks after ART initiation. Cytokine plasma levels were evaluated by Luminex technology at the same time points. The capacity of MDSC to modulate hematopoietic early progenitor cells' expansion was evaluated using the OP9/Dl1 in vitro system. As previously described, polymorphonuclear-MDSC (PMN-MDSC) frequency was higher in PHI compared to healthy donors. Interestingly, 48 weeks of successful ART failed to normalize the PMN-MDSC frequency. Moreover, PMN-MDSC frequency was not correlated with residual viral load, suggesting that the persistence of PMN-MDSC was not due to residual viral replication. Interestingly, patients with low PMN-MDSC frequency (<6%) at T0 had a higher HIV DNA at the same time point than individuals with high PMN-MDSC frequency (>6%). We also found an inverse correlation between PMN-MDSC frequency and CD4-T cell count at 48 weeks post-ART, which was confirmed by multivariate analysis adjusting for age and CD4 T cell number at baseline. These data suggest that the persistence of PMN-MDSC may impact CD4 T cell recovery. Indeed, in vitro PMN-MDSC impaired the expansion of CD34+CD38- hematopoietic early progenitors. Further, a balance between TRAIL and GM-CSF may be necessary to maintain a low MDSC level. In conclusion, early ART initiation was not able to normalize PMN-MDSC frequency that might impact the CD4 T cell recovery. These data open new questions regarding the clinical impact of MDSC persistence in HIV+ patients, in particular on non-AIDS related diseases.
Collapse
Affiliation(s)
- Chiara Agrati
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Nicola Tumino
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Veronica Bordoni
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Carmela Pinnetti
- Virology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Andrea Sabatini
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Alessandra Amendola
- Virology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Isabella Abbate
- Virology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Patrizia Lorenzini
- Clinical Division, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Annalisa Mondi
- Clinical Division, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Rita Casetti
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Eleonora Cimini
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Germana Grassi
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Andrea Antinori
- Clinical Division, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Alessandra Sacchi
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| |
Collapse
|
25
|
Sheng X, Zhang H, Ge P, Chen L, Zhang S. A Retrospective Study of The Prognostic Significance of Preoperative Plasma Fibrinogen, Mean Platelet Volume, and the Neutrophil-to-Lymphocyte Ratio in Patients with Laryngeal Squamous Cell Carcinoma. Med Sci Monit 2019; 25:4527-4534. [PMID: 31210172 PMCID: PMC6597139 DOI: 10.12659/msm.914426] [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] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND This study aimed to evaluate the prognostic significance of plasma fibrinogen, serum albumin, the mean platelet volume (MPV), and the neutrophil-to-lymphocyte ratio (NLR) in patients with laryngeal squamous cell carcinoma (LSCC) who underwent surgical resection. MATERIAL AND METHODS A retrospective study included 110 patients with LSCC who underwent surgical resection between January 2008 to June 2015. Clinicopathologic and demographic data were recorded. Preoperative levels of plasma fibrinogen, serum albumin, MPV, and NLR were measured, and all patients underwent postoperative follow-up. The Kaplan-Meier method was used to determine the impact of these factors on overall survival (OS) and disease-free survival (DFS). RESULTS Preoperative hyperfibrinogenemia was significantly correlated with clinical stage, T stage, and tumor location in patients with LSCC (P<0.05). Serum albumin, MPV, and NLR were significantly correlated with the clinical stage and the T stage (P<0.05). The OS and DFS were significantly reduced in patients with hyperfibrinogenemia compared with patients with plasma fibrinogen <4 g/dL (P<0.05). Serum albumin of 35 g/L was not significantly correlated with OS (P>0.05). Patients with an MPV <9.5 fL had a significantly longer OS compared with patients with an MPV ≥9.5 fL (P=0.026). The DFS of patients with an NLR <2.22 was significantly longer than for those with an NLR ≥2.22. CONCLUSIONS Preoperative hyperfibrinogenemia, increased MPV and NLR were associated with reduced prognosis in patients with LSCC.
Collapse
Affiliation(s)
- Xiaoli Sheng
- Department of Otorhinolaryngology, Guangdong Provincial Peoples' Hospital and Guangdong Acadamy of Medical Sciences, Guangzhou, Guangdong, China (mainland).,Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Heng Zhang
- State Key Laboratory of Respiratory Disease, Department of Pathology, First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China (mainland)
| | - Pingjiang Ge
- Department of Otorhinolaryngology, Guangdong Provincial Peoples' Hospital and Guangdong Acadamy of Medical Sciences, Guangzhou, Guangdong, China (mainland).,Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Liangsi Chen
- Department of Otorhinolaryngology, Guangdong Provincial Peoples' Hospital and Guangdong Acadamy of Medical Sciences, Guangzhou, Guangdong, China (mainland).,Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Siyi Zhang
- Department of Otorhinolaryngology, Guangdong Provincial Peoples' Hospital and Guangdong Acadamy of Medical Sciences, Guangzhou, Guangdong, China (mainland).,Southern Medical University, Guangzhou, Guangdong, China (mainland)
| |
Collapse
|
26
|
Liu H, Zou M, Li P, Wang H, Lin X, Ye J. Oxymatrine‑mediated maturation of dendritic cells leads to activation of FOXP3+/CD4+ Treg cells and reversal of cisplatin‑resistance in lung cancer cells. Mol Med Rep 2019; 19:4081-4090. [PMID: 30896871 PMCID: PMC6471056 DOI: 10.3892/mmr.2019.10064] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 01/14/2019] [Indexed: 12/12/2022] Open
Abstract
The dendritic cell (DC)‑regulatory T (Treg) system serves a leading role in the immunosuppression of the tumor microenvironment, which is not conducive to radiotherapy and chemotherapy treatment for lung cancer. The present study aimed to investigate the effect of oxymatrine (OMT) on the DC‑Treg system in the tumor microenvironment in vitro and to examine its mechanism. The expressions of CD83 antigen, T‑lymphocyte activation antigen CD86, CD11 antigen‑like family member C and major histocompatibility complex II in DCs were increased upon treatment with 1 mg/ml OMT, as detected by flow cytometry. Following pretreatment with OMT, the DCs mediated the forkhead box protein P3 overexpression in primitive cluster of differentiation 4+ T cells at the protein and mRNA expression levels. The expression levels of anti‑inflammatory factors, including interleukin (IL)‑10, tumor growth factor‑β, IL‑35, and pro‑inflammatory cytokines, including interferon‑γ, IL‑12 and IL‑2, in the co‑culture supernatant were increased as measured by ELISA. When DCs and DC‑Tregs were co‑cultured with cisplatin‑resistant A549 cells, the proportion of apoptosis in the co‑culture groups was increased under treatment with cisplatin, which was detected by Annexin V/propidium Iodide staining and western blotting. The present results suggested that OMT may promote the maturation of DCs, mediate the differentiation of T cells into Treg cells, and reverse the resistance of tumor cells to cisplatin in vitro. It was suggested that OMT is an important adjunct to chemotherapy through the regulation of antitumor responses.
Collapse
Affiliation(s)
- Hui Liu
- Division of Pulmonary and Critical Care, Department of Internal Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 501630, P.R. China
| | - Manman Zou
- Division of Pulmonary and Critical Care, Department of Internal Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 501630, P.R. China
| | - Pei Li
- Department of Otolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 501630, P.R. China
| | - Haifeng Wang
- Department of Otolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 501630, P.R. China
| | - Xijun Lin
- Department of Otolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 501630, P.R. China
| | - Jin Ye
- Department of Otolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 501630, P.R. China
| |
Collapse
|
27
|
Abstract
Alpha-fetoprotein is a shuttle protein that delivers nutrients through receptor-mediated endocytosis to embryotic cells. In adults, alpha-fetoprotein can shuttle drugs into alpha-fetoprotein receptor-positive myeloid-derived suppressor, regenerating and also cancer cells. Drugs with high-binding affinity to alpha-fetoprotein can activate or deplete targeted cells. Myeloid-derived suppressor cells activation leads to immune suppression that can be used for treating autoimmune diseases. On the other hand, toxins delivered by alpha-fetoprotein can damage myeloid-derived suppressor cells and consequently unleash innate and adaptive immunity to destroy cancer cells. Innate immunity natural killers reduce cancer stem cells and metastases. The new alpha-fetoprotein drug noncovalent complexes for immunotherapy change the local immune balance and has potential in oncology, autoimmune and infectious diseases treatment, inflammation, transplantation, vaccination, etc.
Collapse
|
28
|
Abstract
In addition to the well-characterized role of AMPK in the regulation of nutrient metabolism, it is increasingly clear that AMPK activation has multiple actions on inflammatory signalling. Here we describe methods to identify effects of AMPK activity on pro-inflammatory signalling, specifically (1) the nuclear localization of the key inflammatory mediators nuclear factor-κB (NFκB) and phosphorylated c-Jun N-terminal kinase (JNK), (2) preparation of conditioned medium to analyze the secretion of cytokines/chemokines, and (3) the pro-inflammatory adhesion of leukocytes to cultured cells.
Collapse
Affiliation(s)
- Sarah J Mancini
- Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Ian P Salt
- Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
29
|
Liu FT, Gao H, Wu CW, Zhu ZM. The association of plasma fibrinogen with clinicopathological features and prognosis in esophageal cancer patients. Oncotarget 2017; 8:93029-93038. [PMID: 29190975 PMCID: PMC5696241 DOI: 10.18632/oncotarget.21746] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 08/04/2017] [Indexed: 01/17/2023] Open
Abstract
Background Numerous studies have shown that plasma fibrinogen was linked to esophageal cancer (EC) risk. However, the clinical significance of plasma fibrinogen in EC patients remain unclear and need to be further clarified. Results A total of 2865 patients with EC from 11 published studies were included in this meta-analysis. The prognostic and clinical relevance of plasma fibrinogen were evaluated in EC patients. Statistical significance of the pooled hazard ratio (HR) was found for overall survival (OS), disease free survival (DFS) and recurrence-free survival (RFS) in EC. Subgroup analyses for OS were also performed to confirm the prognostic value of plasma fibrinogen. Additionally, the overall results indicated that elevated plasma fibrinogen was significantly associated with tumor invasion, lymph node metastasis (LNM) and clinical stage. Materials and Methods A comprehensive literature retrieval was performed in PubMed, Embase, Cochrane database, Web of science and Chinese National Knowledge Infrastructure (CNKI) and Wanfang databases to identify relevant studies published prior to April 15, 2017. Conclusions Elevated plasma fibrinogen could be served as a promising biomarker for predicting a poor prognosis and unfavorable clinicopathologic features for EC.
Collapse
Affiliation(s)
- Fang-Teng Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, P.R. China
| | - Hui Gao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, P.R. China
| | - Chang-Wen Wu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, P.R. China
| | - Zheng-Ming Zhu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, P.R. China
| |
Collapse
|
30
|
Tang CP, Zhou HJ, Qin J, Luo Y, Zhang T. MicroRNA-520c-3p negatively regulates EMT by targeting IL-8 to suppress the invasion and migration of breast cancer. Oncol Rep 2017; 38:3144-3152. [PMID: 29048659 DOI: 10.3892/or.2017.5968] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 08/25/2017] [Indexed: 11/06/2022] Open
Abstract
Interleukin-8 (IL-8), which is secreted by cancer cells undergoing epithelial-mesenchymal transition (EMT), can promote EMT in adjacent epithelial-like cells. MicroRNAs (miRNAs/miRs) can affect the expression of target genes via binding to their 3'-untranslated regions (3'-UTRs), which may subsequently affect the biological behaviors of cancer cells. In our previous study, miR-520c-3p was predicted to directly target the 3'-UTR of IL-8. Therefore, the present study was carried out to investigate whether miR-520c-3p can interact with the IL-8 gene and regulate the EMT of breast cancer cells. Web-based prediction algorithms were used to identify miRNAs that potentially target the IL-8 transcript. Luciferase reporter assays were used to confirm the targeting of IL-8 by miR-520c-3p. Reverse transcription-quantitative PCR and western blot analyses were used to examine the levels of IL-8 and EMT-related genes in breast cancer cells. The functional impact of miR-520c-3p on EMT phenotype was evaluated using Transwell and wound-healing assays, and rescue experiments were conducted by overexpressing IL-8 to determine its effect on cell properties. miR-520c-3p was predicted by all three databases, which strongly suggested its interaction with the 3'-UTR of IL-8. The relative Renilla luciferase activity of luciferase reporter construct containing the wild-type 3'-UTR of IL-8 was markedly decreased by miR-520c-3p transfection when compared with scrambled miRNA control transfection (P<0.001). In addition, compared with the scrambled miRNA control transfection, the overexpression of miR-520c-3p significantly reduced the expression of IL-8, and resulted in increased E-cadherin and decreased vimentin and fibronectin levels in MCF-7 and T47D cells (all P<0.001). Introduction of miR-520c-3p inhibited the invasion and migration of MCF-7 and T47D cells (all P<0.001). By contrast, the rescue of IL-8 expression led to the recovery of EMT-related protein expression patterns and cell motility and invasion capabilities. In conclusion, aberrant miR-520c-3p expression may lead to reduced IL-8 expression and promote the mesenchymal phenotype in breast cancer cells, thereby increasing invasive growth.
Collapse
Affiliation(s)
- Cui-Ping Tang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Han-Jing Zhou
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Jian Qin
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Yi Luo
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Tao Zhang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| |
Collapse
|