1
|
Yoon JH, Bae E, Nagafuchi Y, Sudo K, Han JS, Park SH, Nakae S, Yamashita T, Ju JH, Matsumoto I, Sumida T, Miyazawa K, Kato M, Kuroda M, Lee IK, Fujio K, Mamura M. Repression of SMAD3 by STAT3 and c-Ski induces conventional dendritic cell differentiation. Life Sci Alliance 2024; 7:e201900581. [PMID: 38960622 PMCID: PMC11222659 DOI: 10.26508/lsa.201900581] [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: 10/21/2019] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/05/2024] Open
Abstract
A pleiotropic immunoregulatory cytokine, TGF-β, signals via the receptor-regulated SMADs: SMAD2 and SMAD3, which are constitutively expressed in normal cells. Here, we show that selective repression of SMAD3 induces cDC differentiation from the CD115+ common DC progenitor (CDP). SMAD3 was expressed in haematopoietic cells including the macrophage DC progenitor. However, SMAD3 was specifically down-regulated in CD115+ CDPs, SiglecH- pre-DCs, and cDCs, whereas SMAD2 remained constitutive. SMAD3-deficient mice showed a significant increase in cDCs, SiglecH- pre-DCs, and CD115+ CDPs compared with the littermate control. SMAD3 repressed the mRNA expression of FLT3 and the cDC-related genes: IRF4 and ID2. We found that one of the SMAD transcriptional corepressors, c-SKI, cooperated with phosphorylated STAT3 at Y705 and S727 to repress the transcription of SMAD3 to induce cDC differentiation. These data indicate that STAT3 and c-Ski induce cDC differentiation by repressing SMAD3: the repressor of the cDC-related genes during the developmental stage between the macrophage DC progenitor and CD115+ CDP.
Collapse
Affiliation(s)
- Jeong-Hwan Yoon
- Biomedical Research Institute, Kyungpook National University Hospital, Daegu, Republic of Korea
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
- Shin-Young Medical Institute, Chiba, Japan
- Institute for the 3Rs, Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Eunjin Bae
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
- Department of Companion Health, Yeonsung University, Anyang, Republic of Korea
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yasuo Nagafuchi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Katsuko Sudo
- Animal Research Center, Tokyo Medical University, Tokyo, Japan
| | - Jin Soo Han
- Institute for the 3Rs, Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Seok Hee Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | - Susumu Nakae
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Tadashi Yamashita
- Laboratory of Veterinary Biochemistry, Azabu University School of Veterinary Medicine, Sagamihara, Japan
| | - Ji Hyeon Ju
- Department of Rheumatology, Catholic University of Korea, Seoul St. Mary Hospital, Seoul, Republic of Korea
| | - Isao Matsumoto
- Department of Internal Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takayuki Sumida
- Department of Internal Medicine, University of Tsukuba, Tsukuba, Japan
| | - Keiji Miyazawa
- Departments of Biochemistry, University of Yamanashi, Yamanashi, Japan
| | - Mitsuyasu Kato
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Masahiko Kuroda
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| | - In-Kyu Lee
- Biomedical Research Institute, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mizuko Mamura
- Biomedical Research Institute, Kyungpook National University Hospital, Daegu, Republic of Korea
- Shin-Young Medical Institute, Chiba, Japan
- Department of Advanced Nucleic Acid Medicine, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
2
|
Thenuwara G, Javed B, Singh B, Tian F. Biosensor-Enhanced Organ-on-a-Chip Models for Investigating Glioblastoma Tumor Microenvironment Dynamics. SENSORS (BASEL, SWITZERLAND) 2024; 24:2865. [PMID: 38732975 PMCID: PMC11086276 DOI: 10.3390/s24092865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/19/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024]
Abstract
Glioblastoma, an aggressive primary brain tumor, poses a significant challenge owing to its dynamic and intricate tumor microenvironment. This review investigates the innovative integration of biosensor-enhanced organ-on-a-chip (OOC) models as a novel strategy for an in-depth exploration of glioblastoma tumor microenvironment dynamics. In recent years, the transformative approach of incorporating biosensors into OOC platforms has enabled real-time monitoring and analysis of cellular behaviors within a controlled microenvironment. Conventional in vitro and in vivo models exhibit inherent limitations in accurately replicating the complex nature of glioblastoma progression. This review addresses the existing research gap by pioneering the integration of biosensor-enhanced OOC models, providing a comprehensive platform for investigating glioblastoma tumor microenvironment dynamics. The applications of this combined approach in studying glioblastoma dynamics are critically scrutinized, emphasizing its potential to bridge the gap between simplistic models and the intricate in vivo conditions. Furthermore, the article discusses the implications of biosensor-enhanced OOC models in elucidating the dynamic features of the tumor microenvironment, encompassing cell migration, proliferation, and interactions. By furnishing real-time insights, these models significantly contribute to unraveling the complex biology of glioblastoma, thereby influencing the development of more accurate diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Gayathree Thenuwara
- School of Food Science and Environmental Health, Technological University Dublin, Grangegorman Lower, D07 H6K8 Dublin, Ireland; (G.T.); (B.J.)
- Institute of Biochemistry, Molecular Biology, and Biotechnology, University of Colombo, Colombo 00300, Sri Lanka
| | - Bilal Javed
- School of Food Science and Environmental Health, Technological University Dublin, Grangegorman Lower, D07 H6K8 Dublin, Ireland; (G.T.); (B.J.)
- Nanolab Research Centre, FOCAS Research Institute, Technological University Dublin, Camden Row, D08 CKP1 Dublin, Ireland
| | - Baljit Singh
- MiCRA Biodiagnostics Technology Gateway, Technological University Dublin (TU Dublin), D24 FKT9 Dublin, Ireland;
| | - Furong Tian
- School of Food Science and Environmental Health, Technological University Dublin, Grangegorman Lower, D07 H6K8 Dublin, Ireland; (G.T.); (B.J.)
- Nanolab Research Centre, FOCAS Research Institute, Technological University Dublin, Camden Row, D08 CKP1 Dublin, Ireland
| |
Collapse
|
3
|
Elguindy M, Young JS, Mondal I, Lu RO, Ho WS. Glioma-Immune Cell Crosstalk in Tumor Progression. Cancers (Basel) 2024; 16:308. [PMID: 38254796 PMCID: PMC10813573 DOI: 10.3390/cancers16020308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/21/2023] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
Glioma progression is a complex process controlled by molecular factors that coordinate the crosstalk between tumor cells and components of the tumor microenvironment (TME). Among these, immune cells play a critical role in cancer survival and progression. The complex interplay between cancer cells and the immune TME influences the outcome of immunotherapy and other anti-cancer therapies. Here, we present an updated view of the pro- and anti-tumor activities of the main myeloid and lymphocyte cell populations in the glioma TME. We review the underlying mechanisms involved in crosstalk between cancer cells and immune cells that enable gliomas to evade the immune system and co-opt these cells for tumor growth. Lastly, we discuss the current and experimental therapeutic options being developed to revert the immunosuppressive activity of the glioma TME. Knowledge of the complex interplay that elapses between tumor and immune cells may help develop new combination treatments able to overcome tumor immune evasion mechanisms and enhance response to immunotherapies.
Collapse
Affiliation(s)
| | | | | | | | - Winson S. Ho
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
4
|
Halasi M, Talmon A, Tal Y, Yosipovitch G, Adini I. Dark pigmentation and related low FMOD expression increase IL-3 and facilitate plasmacytoid dendritic cell maturation. Clin Immunol 2023; 251:109638. [PMID: 37149118 DOI: 10.1016/j.clim.2023.109638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 05/08/2023]
Abstract
According to epidemiological research, skin autoimmune diseases are more prevalent among black Americans. We postulated that pigment-producing melanocytes may contribute to local immune regulation in the microenvironment. We examined murine epidermal melanocytes in vitro to determine the role of pigment production in immune responses mediated by dendritic cell (DC) activation. Our study revealed that darkly pigmented melanocytes produce more IL-3 and the pro-inflammatory cytokines, IL-6 and TNF-α, and consequently induce plasmacytoid DC (pDC) maturation. Additionally, we demonstrate that low pigment-associated fibromodulin (FMOD) interferes with cytokine secretion and subsequent pDC maturation.
Collapse
Affiliation(s)
- Marianna Halasi
- Harvard Medical School, Department of Surgery, Center for Engineering in Medicine & Surgery, Massachusetts General Hospital, 51 Blossom Street, Boston, MA 02114, United States of America
| | - Aviv Talmon
- Allergy and Clinical Immunology Unit, Department of Medicine, Hadassah Medical Organization, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Yuval Tal
- Allergy and Clinical Immunology Unit, Department of Medicine, Hadassah Medical Organization, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Gil Yosipovitch
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery and Miami Itch Ctr, University of Miami, FL, USA
| | - Irit Adini
- Harvard Medical School, Department of Surgery, Center for Engineering in Medicine & Surgery, Massachusetts General Hospital, 51 Blossom Street, Boston, MA 02114, United States of America.
| |
Collapse
|
5
|
Decraene B, Vanmechelen M, Clement P, Daisne JF, Vanden Bempt I, Sciot R, Garg AD, Agostinis P, De Smet F, De Vleeschouwer S. Cellular and molecular features related to exceptional therapy response and extreme long-term survival in glioblastoma. Cancer Med 2023. [PMID: 36776000 DOI: 10.1002/cam4.5681] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/17/2023] [Accepted: 01/31/2023] [Indexed: 02/14/2023] Open
Abstract
Glioblastoma Multiforme (GBM) remains the most common malignant primary brain tumor with a dismal prognosis that rarely exceeds beyond 2 years despite extensive therapy, which consists of maximal safe surgical resection, radiotherapy, and/or chemotherapy. Recently, it has become clear that GBM is not one homogeneous entity and that both intra-and intertumoral heterogeneity contributes significantly to differences in tumoral behavior which may consequently be responsible for differences in survival. Strikingly and in spite of its dismal prognosis, small fractions of GBM patients seem to display extremely long survival, defined as surviving over 10 years after diagnosis, compared to the large majority of patients. Although the underlying mechanisms for this peculiarity remain largely unknown, emerging data suggest that still poorly characterized both cellular and molecular factors of the tumor microenvironment and their interplay probably play an important role. We hereby give an extensive overview of what is yet known about these cellular and molecular features shaping extreme long survival in GBM.
Collapse
Affiliation(s)
- B Decraene
- KU Leuven, Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Leuven, Belgium.,KU Leuven Department of Neurosciences, Experimental Neurosurgery and Neuroanatomy Research Group, Leuven, Belgium.,Department of Neurosurgery, University Hospitals Leuven, Leuven, Belgium
| | - M Vanmechelen
- KU Leuven, Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Leuven, Belgium.,Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - P Clement
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - J F Daisne
- Radiation Oncology Department, University Hospitals Leuven, Leuven, Belgium
| | - I Vanden Bempt
- Department of Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - R Sciot
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - A D Garg
- KU Leuven, VIB Center for Cancer Biology Research, Leuven, Belgium
| | - P Agostinis
- KU Leuven, Laboratory of Cell Stress & Immunity (CSI), Department of Cellular & Molecular Medicine, Leuven, Belgium
| | - F De Smet
- KU Leuven, Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Leuven, Belgium
| | - S De Vleeschouwer
- KU Leuven Department of Neurosciences, Experimental Neurosurgery and Neuroanatomy Research Group, Leuven, Belgium.,Department of Neurosurgery, University Hospitals Leuven, Leuven, Belgium.,KU Leuven, Leuven Brain Institute (LBI), Leuven, Belgium
| |
Collapse
|
6
|
The Tumor Immune Microenvironment in Primary CNS Neoplasms: A Review of Current Knowledge and Therapeutic Approaches. Int J Mol Sci 2023; 24:ijms24032020. [PMID: 36768342 PMCID: PMC9917056 DOI: 10.3390/ijms24032020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Primary CNS neoplasms are responsible for considerable mortality and morbidity, and many therapies directed at primary brain tumors have proven unsuccessful despite their success in preclinical studies. Recently, the tumor immune microenvironment has emerged as a critical aspect of primary CNS neoplasms that may affect their malignancy, prognosis, and response to therapy across patients and tumor grades. This review covers the tumor microenvironment of various primary CNS neoplasms, with a focus on glioblastoma and meningioma. Additionally, current therapeutic strategies based on elements of the tumor microenvironment, including checkpoint inhibitor therapy and immunotherapeutic vaccines, are discussed.
Collapse
|
7
|
Piacente F, Bottero M, Benzi A, Vigo T, Uccelli A, Bruzzone S, Ferrara G. Neuroprotective Potential of Dendritic Cells and Sirtuins in Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms23084352. [PMID: 35457169 PMCID: PMC9025744 DOI: 10.3390/ijms23084352] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/06/2022] [Accepted: 04/11/2022] [Indexed: 12/04/2022] Open
Abstract
Myeloid cells, including parenchymal microglia, perivascular and meningeal macrophages, and dendritic cells (DCs), are present in the central nervous system (CNS) and establish an intricate relationship with other cells, playing a crucial role both in health and in neurological diseases. In this context, DCs are critical to orchestrating the immune response linking the innate and adaptive immune systems. Under steady-state conditions, DCs patrol the CNS, sampling their local environment and acting as sentinels. During neuroinflammation, the resulting activation of DCs is a critical step that drives the inflammatory response or the resolution of inflammation with the participation of different cell types of the immune system (macrophages, mast cells, T and B lymphocytes), resident cells of the CNS and soluble factors. Although the importance of DCs is clearly recognized, their exact function in CNS disease is still debated. In this review, we will discuss modern concepts of DC biology in steady-state and during autoimmune neuroinflammation. Here, we will also address some key aspects involving DCs in CNS patrolling, highlighting the neuroprotective nature of DCs and emphasizing their therapeutic potential for the treatment of neurological conditions. Recently, inhibition of the NAD+-dependent deac(et)ylase sirtuin 6 was demonstrated to delay the onset of experimental autoimmune encephalomyelitis, by dampening DC trafficking towards inflamed LNs. Thus, a special focus will be dedicated to sirtuins’ role in DCs functions.
Collapse
Affiliation(s)
- Francesco Piacente
- Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (A.B.)
| | - Marta Bottero
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| | - Andrea Benzi
- Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (A.B.)
| | - Tiziana Vigo
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| | - Antonio Uccelli
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| | - Santina Bruzzone
- Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (A.B.)
- Correspondence: ; Tel.: +39-(0)10-353-8150
| | - Giovanni Ferrara
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| |
Collapse
|
8
|
Kharbanda A, Tran P, Zhang L, Leung YK, Li HY, Frett B. Discovery of 4-aminoquinolines as highly selective TGFβR1 inhibitors with an attenuated MAP4K4 profile for potential applications in immuno-oncology. Eur J Med Chem 2021; 225:113763. [PMID: 34419892 DOI: 10.1016/j.ejmech.2021.113763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 11/19/2022]
Abstract
The tumor microenvironment contains high concentrations of TGFβ, a crucial immunosuppressive cytokine. TGFβ stimulates immune escape by promoting peripheral immune tolerance to avoid tumoricidal attack. Small-molecule inhibitors of TGFβR1 are a prospective method for next-generation immunotherapies. In the present study, we identified selective 4-aminoquinoline-based inhibitors of TGFβR1 through structural and rational-based design strategies. This led to the identification of compound 4i, which was found to be selective for TGFβR1 with the exception of MAP4K4 in the kinase profiling assay. The compound was then further optimized to remove MAP4K4 activity, since MAP4K4 is vital for proper T-cell function and its inhibition could exacerbate tumor immunosuppression. Optimization efforts led to compound 4s that inhibited TGFβR1 at an IC50 of 0.79 ± 0.19 nM with 2000-fold selectivity against MAP4K4. Compound 4s represents a highly selective TGFβR1 inhibitor that has potential applications in immuno-oncology.
Collapse
Affiliation(s)
- Anupreet Kharbanda
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Phuc Tran
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Lingtian Zhang
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yuet-Kin Leung
- Department of Pharmacology & Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Hong-Yu Li
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| | - Brendan Frett
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
9
|
Fisetin Attenuates Lipopolysaccharide-Induced Inflammatory Responses in Macrophage. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5570885. [PMID: 33954178 PMCID: PMC8057890 DOI: 10.1155/2021/5570885] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/23/2021] [Accepted: 03/31/2021] [Indexed: 12/30/2022]
Abstract
Several studies have reported the efficacy and safety of polyphenols in human health; however, the verification of their efficacy remains insufficient. The aim of this study was to examine whether fisetin, one of flavonoids prevalently present in fruits and vegetables, could suppress lipopolysaccharide- (LPS-) induced inflammatory responses in macrophages. LPS increased proinflammatory mRNA abundance (MCP 1, IL-1β, and iNOS) but were suppressed by fisetin. The increment of nitric oxide by LPS, an oxidative stress factor, was attenuated by fisetin. In addition, LPS-enhanced phosphorylation of mitogen-activated protein kinase (ERK and JNK) was reduced. Finally, fisetin attenuated the expression or activity of uPA, uPAR, MMP-2, and MMP-9, which are known as associated factors of macrophage recruitment or infiltration. In conclusion, fisetin is a promising therapeutic agent for macrophage-related inflammation diseases, like sepsis and atherosclerosis.
Collapse
|
10
|
Chen J, Ding ZY, Li S, Liu S, Xiao C, Li Z, Zhang BX, Chen XP, Yang X. Targeting transforming growth factor-β signaling for enhanced cancer chemotherapy. Theranostics 2021; 11:1345-1363. [PMID: 33391538 PMCID: PMC7738904 DOI: 10.7150/thno.51383] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/29/2020] [Indexed: 12/14/2022] Open
Abstract
During the past decades, drugs targeting transforming growth factor-β (TGFβ) signaling have received tremendous attention for late-stage cancer treatment since TGFβ signaling has been recognized as a prime driver for tumor progression and metastasis. Nonetheless, in healthy and pre-malignant tissues, TGFβ functions as a potent tumor suppressor. Furthermore, TGFβ signaling plays a key role in normal development and homeostasis by regulating cell proliferation, differentiation, migration, apoptosis, and immune evasion, and by suppressing tumor-associated inflammation. Therefore, targeting TGFβ signaling for cancer therapy is challenging. Recently, we and others showed that blocking TGFβ signaling increased chemotherapy efficacy, particularly for nanomedicines. In this review, we briefly introduce the TGFβ signaling pathway, and the multifaceted functions of TGFβ signaling in cancer, including regulating the tumor microenvironment (TME) and the behavior of cancer cells. We also summarize TGFβ targeting agents. Then, we highlight TGFβ inhibition strategies to restore the extracellular matrix (ECM), regulate the tumor vasculature, reverse epithelial-mesenchymal transition (EMT), and impair the stemness of cancer stem-like cells (CSCs) to enhance cancer chemotherapy efficacy. Finally, the current challenges and future opportunities in targeting TGFβ signaling for cancer therapy are discussed.
Collapse
Affiliation(s)
- Jitang Chen
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Ze-yang Ding
- Hepatic Surgery Center, and Hubei Key Laboratory of Hepatic-Biliary-Pancreatic Diseases, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Si Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Sha Liu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Hepatic Surgery Center, and Hubei Key Laboratory of Hepatic-Biliary-Pancreatic Diseases, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Xiao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zifu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Bi-xiang Zhang
- Hepatic Surgery Center, and Hubei Key Laboratory of Hepatic-Biliary-Pancreatic Diseases, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-ping Chen
- Hepatic Surgery Center, and Hubei Key Laboratory of Hepatic-Biliary-Pancreatic Diseases, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan, 430074, China
- GBA Research Innovation Institute for Nanotechnology, Guangdong, 510530, China
| |
Collapse
|
11
|
Thomson AW, Vionnet J, Sanchez-Fueyo A. Understanding, predicting and achieving liver transplant tolerance: from bench to bedside. Nat Rev Gastroenterol Hepatol 2020; 17:719-739. [PMID: 32759983 DOI: 10.1038/s41575-020-0334-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/11/2020] [Indexed: 02/07/2023]
Abstract
In the past 40 years, liver transplantation has evolved from a high-risk procedure to one that offers high success rates for reversal of liver dysfunction and excellent patient and graft survival. The liver is the most tolerogenic of transplanted organs; indeed, immunosuppressive therapy can be completely withdrawn without rejection of the graft in carefully selected, stable long-term liver recipients. However, in other recipients, chronic allograft injury, late graft failure and the adverse effects of anti-rejection therapy remain important obstacles to improved success. The liver has a unique composition of parenchymal and immune cells that regulate innate and adaptive immunity and that can promote antigen-specific tolerance. Although the mechanisms underlying liver transplant tolerance are not well understood, important insights have been gained into how the local microenvironment, hepatic immune cells and specific molecular pathways can promote donor-specific tolerance. These insights provide a basis for the identification of potential clinical biomarkers that might correlate with tolerance or rejection and for the development of novel therapeutic targets. Innovative approaches aimed at promoting immunosuppressive drug minimization or withdrawal include the adoptive transfer of donor-derived or recipient-derived regulatory immune cells to promote liver transplant tolerance. In this Review, we summarize and discuss these developments and their implications for liver transplantation.
Collapse
Affiliation(s)
- Angus W Thomson
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. .,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Julien Vionnet
- Institute of Liver Studies, Medical Research Council (MRC) Centre for Transplantation, School of Immunology and Infectious Diseases, King's College London University, King's College Hospital, London, UK.,Transplantation Center, University Hospital of Lausanne, Lausanne, Switzerland.,Service of Gastroenterology and Hepatology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Alberto Sanchez-Fueyo
- Institute of Liver Studies, Medical Research Council (MRC) Centre for Transplantation, School of Immunology and Infectious Diseases, King's College London University, King's College Hospital, London, UK
| |
Collapse
|
12
|
Nejo T, Mende A, Okada H. The current state of immunotherapy for primary and secondary brain tumors: similarities and differences. Jpn J Clin Oncol 2020; 50:1231-1245. [PMID: 32984905 DOI: 10.1093/jjco/hyaa164] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/11/2020] [Indexed: 12/14/2022] Open
Abstract
Treatment and resolution of primary and metastatic brain tumors have long presented a challenge to oncologists. In response to the dismal survival outcomes associated with conventional therapies, various immunotherapy modalities, such as checkpoint inhibitors, vaccine, cellular immunotherapy and viral immunotherapy have been actively explored over the past couple of decades. Although improved patient survival has been more frequently noted in treatment of brain metastases, little progress has been made in improving patient survival in cases of primary brain tumors, specifically glioblastoma, which is the representative primary brain tumor discussed in this review. Herein, we will first overview the findings of recent clinical studies for treatment of primary and metastatic brain tumors with immunotherapeutic interventions. The clinical efficacy of these immunotherapies will be discussed in the context of their ability or inability to overcome inherent characteristics of the tumor as well as restricted antigen presentation and its immunosuppressive microenvironment. Additionally, this review aims to briefly inform clinicians in the field of neuro-oncology on the relevant aspects of the immune system as it pertains to the central nervous system, with special focus on the differing modes of antigen presentation and tumor microenvironment of primary and metastatic brain tumors and the role these differences may play in the efficacy of immunotherapy in eradicating the tumor.
Collapse
Affiliation(s)
- Takahide Nejo
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Abigail Mende
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Hideho Okada
- Department of Neurological Surgery, University of California, San Francisco, CA, USA.,The Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.,Cancer Immunotherapy Program, University of California, San Francisco, CA, USA
| |
Collapse
|
13
|
Somri-Gannam L, Meisel-Sharon S, Hantisteanu S, Groisman G, Limonad O, Hallak M, Bruchim I. IGF1R Axis Inhibition Restores Dendritic Cell Antitumor Response in Ovarian Cancer. Transl Oncol 2020; 13:100790. [PMID: 32428851 PMCID: PMC7232112 DOI: 10.1016/j.tranon.2020.100790] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/24/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecological malignancy. The insulin-like growth factor (IGF) system plays a key role in regulating growth and invasiveness in several malignancies, including ovarian cancer. IGF1R targeting showed antiproliferative activity of EOC cells. However, clinical studies failed to show significant benefit. EOC cells suppress antitumor immune responses by inducing dendritic cell (DC) dysfunction. The IGF1 axis can regulate DC maturation. The current study evaluated involvement of the IGF1 axis in DC differentiation in EOC. Studies were conducted on EOC and on a human monocyte cell line. Tissue microarray analysis (TMA) was performed on 36 paraffin blocks from EOC patients. Expression of IGF1R, p53, Ki67, BRCA1, and DC markers was evaluated using immunohistochemistry. Co-culture of EOC cells with DC pretreated with IGF1R inhibitor blocked cancer cell migration. TMA demonstrated higher rate of IGF1R protein expression in patients with advanced (76.9%) as compared to early (40%) EOC. A negative correlation between IGF1R protein expression and the CD1c marker was found. These findings provide evidence that IGF1R axis inhibition could be a therapeutic strategy for ovarian cancer by restoring DC-mediated antitumor immunity.
Collapse
Affiliation(s)
- Lina Somri-Gannam
- Gynecology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.
| | - Shilhav Meisel-Sharon
- Gynecology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Israel
| | - Shay Hantisteanu
- Gynecology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Israel
| | - Gabriel Groisman
- Institute of Pathology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Ofer Limonad
- Gynecology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Israel; Gynecologic Oncology Division, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Mordechai Hallak
- Gynecology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel; Gynecologic Oncology Division, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Ilan Bruchim
- Gynecology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel; Gynecologic Oncology Division, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
| |
Collapse
|
14
|
Krop J, Heidt S, Claas FHJ, Eikmans M. Regulatory T Cells in Pregnancy: It Is Not All About FoxP3. Front Immunol 2020; 11:1182. [PMID: 32655556 PMCID: PMC7324675 DOI: 10.3389/fimmu.2020.01182] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/13/2020] [Indexed: 12/15/2022] Open
Abstract
In pregnancy, the semi-allogeneic fetus needs to be tolerated by the mother's immune system. Regulatory T cells (Tregs) play a prominent role in this process. Novel technologies allow for in-depth phenotyping of previously unidentified immune cell subsets, which has resulted in the appreciation of a vast heterogeneity of Treg subsets. Similar to other immunological events, there appears to be great diversity within the Treg population during pregnancy, both at the maternal-fetal interface as in the peripheral blood. Different Treg subsets have distinct phenotypes and various ways of functioning. Furthermore, the frequency of individual Treg subsets varies throughout gestation and is altered in aberrant pregnancies. This suggests that distinct Treg subsets play a role at different time points of gestation and that their role in maintaining healthy pregnancy is crucial, as reflected for instance by their reduced frequency in women with recurrent pregnancy loss. Since pregnancy is essential for the existence of mankind, multiple immune regulatory mechanisms and cell types are likely at play to assure successful pregnancy. Therefore, it is important to understand the complete microenvironment of the decidua, preferably in the context of the whole immune cell repertoire of the pregnant woman. So far, most studies have focused on a single mechanism or cell type, which often is the FoxP3 positive regulatory T cell when studying immune regulation. In this review, we instead focus on the contribution of FoxP3 negative Treg subsets to the decidual microenvironment and their possible role in pregnancy complications. Their phenotype, function, and effect in pregnancy are discussed.
Collapse
Affiliation(s)
- Juliette Krop
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Sebastiaan Heidt
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Frans H J Claas
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Michael Eikmans
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
15
|
Zhang N, Wang Z, Zhao Y. Selective inhibition of Tumor necrosis factor receptor-1 (TNFR1) for the treatment of autoimmune diseases. Cytokine Growth Factor Rev 2020; 55:80-85. [PMID: 32327345 DOI: 10.1016/j.cytogfr.2020.03.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 03/30/2020] [Indexed: 12/19/2022]
Abstract
Anti-TNF biologics have achieved great success in the treatment of autoimmune diseases and have been the most selling biologics on market. However, the anti-TNF biologics have shown some disadvantages such as poor efficacy to some patients and high risk of infection and malignancies during clinical application. Current anti-TNF biologics are antibodies or antibody fragments that bind to TNF-α and subsequently block both TNF-TNFR1 and TNF-TNFR2 signaling. Transgenic animal studies indicate that TNFR1 signaling is responsible for chronic inflammation and cell apoptosis whereas TNFR2 signaling regulates tissue regeneration and inflammation. Recent studies propose to selectively inhibit TNFR1 to enhance efficacy and avoid side effects. In this review, we introduce the biology of TNF-TNFR1 and TNF-TNFR2 signaling, the advantages of selective inhibition of TNF-TNFR1 signaling and research updates on the development of selective inhibitors for TNF-TNFR1 signaling. Antibodies, small molecules and aptamers that selectively inhibit TNFR1 have showed therapeutic potential and less side effects in preclinical studies. Development of selective inhibitors for TNFR1 is a good strategy to enhance the efficacy and reduce the side effects of anti-TNF inhibitors and will be a trend for next-generation of anti-TNF inhibitors.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, PR China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, HeNan Province, Zhengzhou 450001, Henan, PR China; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, HeNan Province, Zhengzhou 450001, Henan, PR China
| | - Ziyi Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, PR China
| | - Yongxing Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, PR China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, HeNan Province, Zhengzhou 450001, Henan, PR China; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, HeNan Province, Zhengzhou 450001, Henan, PR China.
| |
Collapse
|
16
|
Voisin A, Damon-Soubeyrand C, Bravard S, Saez F, Drevet JR, Guiton R. Differential expression and localisation of TGF-β isoforms and receptors in the murine epididymis. Sci Rep 2020; 10:995. [PMID: 31969637 PMCID: PMC6976608 DOI: 10.1038/s41598-020-57839-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 12/16/2019] [Indexed: 12/21/2022] Open
Abstract
Testes produce spermatozoa that transit through and are stored in the epididymis where they acquire their fertilising capacities. Spermatozoa appear in the genital tract at puberty, long after the immune system was trained to self-antigens. As a consequence, this organ has to set strategies to tolerate sperm antigens to avoid autoimmune responses that would specifically target and destroy them. A recent study pointed the Transforming Growth Factor-beta (TGF-β) signalling in the dendritic cells as a crucial mechanism for epididymal tolerance to spermatozoa. In the mouse, TGF-β exists under three isoforms, and three distinct receptors have been described. Using RT-qPCR, immunohistochemistry and ELISA techniques, we investigated the expression and spatial distribution of the epididymal TGF-β isoforms and of their receptors in young and adult mice. We showed that both ligands and receptors were produced by immune and non-immune cells in the epididymis, whatever the age mice have. These data bring new clues as to the mechanisms of peripheral tolerance to sperm cells in the murine epididymis and raise potential other implications of the cytokine isoforms.
Collapse
Affiliation(s)
- Allison Voisin
- GReD laboratory, CNRS UMR 6293 - INSERM U1103 - Université Clermont Auvergne, 28 place Henri Dunant, 63001, Clermont-Ferrand Cedex, France
| | - Christelle Damon-Soubeyrand
- GReD laboratory, CNRS UMR 6293 - INSERM U1103 - Université Clermont Auvergne, 28 place Henri Dunant, 63001, Clermont-Ferrand Cedex, France
| | - Stéphanie Bravard
- GReD laboratory, CNRS UMR 6293 - INSERM U1103 - Université Clermont Auvergne, 28 place Henri Dunant, 63001, Clermont-Ferrand Cedex, France
| | - Fabrice Saez
- GReD laboratory, CNRS UMR 6293 - INSERM U1103 - Université Clermont Auvergne, 28 place Henri Dunant, 63001, Clermont-Ferrand Cedex, France
| | - Joël R Drevet
- GReD laboratory, CNRS UMR 6293 - INSERM U1103 - Université Clermont Auvergne, 28 place Henri Dunant, 63001, Clermont-Ferrand Cedex, France.
| | - Rachel Guiton
- GReD laboratory, CNRS UMR 6293 - INSERM U1103 - Université Clermont Auvergne, 28 place Henri Dunant, 63001, Clermont-Ferrand Cedex, France.
| |
Collapse
|
17
|
Eleuteri S, Fierabracci A. Insights into the Secretome of Mesenchymal Stem Cells and Its Potential Applications. Int J Mol Sci 2019; 20:4597. [PMID: 31533317 PMCID: PMC6770239 DOI: 10.3390/ijms20184597] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/09/2019] [Accepted: 09/11/2019] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have regenerative, immunoregulatory properties and can be easily isolated and expanded in vitro. Despite being a powerful tool for clinical applications, they present limitations in terms of delivery, safety, and variability of therapeutic response. Interestingly, the MSC secretome composed by cytokines, chemokines, growth factors, proteins, and extracellular vesicles, could represent a valid alternative to their use. It is noteworthy that MSC-derived extracellular vesicles (MSC-EVs) have the same effect and could be advantageous compared to the parental cells because of their specific miRNAs load. MiRNAs could be useful both in diagnostic procedures such as "liquid biopsy" to identify early pathologies and in the therapeutic field. Not only are MSC-EVs' preservation, transfer, and production easier, but their administration is also safer, hence some clinical trials are ongoing. However, much effort is required to improve the characterization of EVs to avoid artifacts and guarantee reproducibility of the studies.
Collapse
Affiliation(s)
- Sharon Eleuteri
- Infectivology and Clinical Trials Area, Children's Hospital Bambino Gesù, Viale San Paolo 15, 00146 Rome, Italy.
| | - Alessandra Fierabracci
- Infectivology and Clinical Trials Area, Children's Hospital Bambino Gesù, Viale San Paolo 15, 00146 Rome, Italy.
| |
Collapse
|
18
|
Braun E, Sauter D. Furin-mediated protein processing in infectious diseases and cancer. Clin Transl Immunology 2019; 8:e1073. [PMID: 31406574 PMCID: PMC6682551 DOI: 10.1002/cti2.1073] [Citation(s) in RCA: 217] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/10/2019] [Accepted: 07/13/2019] [Indexed: 12/17/2022] Open
Abstract
Proteolytic cleavage regulates numerous processes in health and disease. One key player is the ubiquitously expressed serine protease furin, which cleaves a plethora of proteins at polybasic recognition motifs. Mammalian substrates of furin include cytokines, hormones, growth factors and receptors. Thus, it is not surprising that aberrant furin activity is associated with a variety of disorders including cancer. Furthermore, the enzymatic activity of furin is exploited by numerous viral and bacterial pathogens, thereby enhancing their virulence and spread. In this review, we describe the physiological and pathophysiological substrates of furin and discuss how dysregulation of a simple proteolytic cleavage event may promote infectious diseases and cancer. One major focus is the role of furin in viral glycoprotein maturation and pathogenicity. We also outline cellular mechanisms regulating the expression and activation of furin and summarise current approaches that target this protease for therapeutic intervention.
Collapse
Affiliation(s)
- Elisabeth Braun
- Institute of Molecular VirologyUlm University Medical CenterUlmGermany
| | - Daniel Sauter
- Institute of Molecular VirologyUlm University Medical CenterUlmGermany
| |
Collapse
|
19
|
Mei XF, Shi W, Zhang YY, Zhu B, Wang YR, Hou LJ, Zhao WP, Li J, Wang DY, Luo HL, Huang WY. DNA methylation and hydroxymethylation profiles reveal possible role of highly methylated TLR signaling on Fasciola gigantica excretory/secretory products (FgESPs) modulation of buffalo dendritic cells. Parasit Vectors 2019; 12:358. [PMID: 31337442 PMCID: PMC6647289 DOI: 10.1186/s13071-019-3615-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 07/13/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Excretory/secretory products (ESPs) released by parasites influence the development and functions of host dendritic cells (DCs). However, little is known about changes of DNA (hydroxy)methylation on DC development during Fasciola gigantica infection. The present study aimed to investigate whether F. gigantica ESPs (FgESPs) affects the development and functions of buffalo DCs through altering the DNA (hydroxy)methylation of DCs. METHODS Buffalo DCs were prepared from peripheral blood mononuclear cells (PBMCs) and characterized using scanning and transmission electron microscopy (SEM/TEM) and quantitative reverse transcriptional PCR (qRT-RCR). DCs were treated with 200 μg/ml of FgESPs in vitro, following DNA extraction. The DNA methylome and hydroxymethylome were profiled based on (hydroxy)methylated DNA immunoprecipitation sequencing [(h)MeDIP-Seq] and bioinformatics analyses. qRT-RCR was also performed to assess the gene transcription levels of interest. RESULTS FgESPs markedly suppressed DC maturation evidenced by morphological changes and downregulated gene expression of CD1a and MHC II. Totals of 5432 and 360 genes with significant changes in the 5-methylcytosine (5-mC) and the 5-hydroxymethylcytosine (5-hmC) levels, respectively, were identified in buffalo DCs in response to FgESPs challenge. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed that these differentially expressed genes were highly enriched in pathways associated with immune response. Some cancer-related pathways were also indicated. There were 111 genes demonstrating changes in both 5-mC and 5-hmC levels, 12 of which were interconnected and enriched in 12 pathways. The transcription of hypermethylated genes TLR2, TLR4 and IL-12B were downregulated or in a decreasing trend, while the mRNA level of high-hydroxymethylated TNF gene was upregulated in buffalo DCs post-exposure to FgESPs in vitro. CONCLUSIONS To our knowledge, the present study provides for the first time a unique genome-wide profile of DNA (hydroxy)methylation for DCs that interact with FgESPs, and suggests a possible mechanism of FgESPs in suppressing DC maturation and functions that are involved in TLR signaling.
Collapse
Affiliation(s)
- Xue-Fang Mei
- School of Animal Science and Technology, Guangxi University, Nanning, People's Republic of China
| | - Wei Shi
- School of Preclinical Medicine, Guangxi Medical University, Nanning, People's Republic of China
| | - Yao-Yao Zhang
- School of Animal Science and Technology, Guangxi University, Nanning, People's Republic of China
| | - Bin Zhu
- School of Animal Science and Technology, Guangxi University, Nanning, People's Republic of China
| | - Yu-Rui Wang
- School of Animal Science and Technology, Guangxi University, Nanning, People's Republic of China
| | - Lin-Jing Hou
- School of Animal Science and Technology, Guangxi University, Nanning, People's Republic of China
| | - Wen-Ping Zhao
- School of Animal Science and Technology, Guangxi University, Nanning, People's Republic of China
| | - Jian Li
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, People's Republic of China
| | - Dong-Ying Wang
- School of Animal Science and Technology, Guangxi University, Nanning, People's Republic of China.
| | - Hong-Lin Luo
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Institute of Fishery Sciences, Nanning, People's Republic of China.
| | - Wei-Yi Huang
- School of Animal Science and Technology, Guangxi University, Nanning, People's Republic of China.
| |
Collapse
|
20
|
Chrisikos TT, Zhou Y, Slone N, Babcock R, Watowich SS, Li HS. Molecular regulation of dendritic cell development and function in homeostasis, inflammation, and cancer. Mol Immunol 2019; 110:24-39. [PMID: 29549977 PMCID: PMC6139080 DOI: 10.1016/j.molimm.2018.01.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 01/04/2018] [Accepted: 01/25/2018] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) are the principal antigen-presenting cells of the immune system and play key roles in controlling immune tolerance and activation. As such, DCs are chief mediators of tumor immunity. DCs can regulate tolerogenic immune responses that facilitate unchecked tumor growth. Importantly, however, DCs also mediate immune-stimulatory activity that restrains tumor progression. For instance, emerging evidence indicates the cDC1 subset has important functions in delivering tumor antigens to lymph nodes and inducing antigen-specific lymphocyte responses to tumors. Moreover, DCs control specific therapeutic responses in cancer including those resulting from immune checkpoint blockade. DC generation and function is influenced profoundly by cytokines, as well as their intracellular signaling proteins including STAT transcription factors. Regardless, our understanding of DC regulation in the cytokine-rich tumor microenvironment is still developing and must be better defined to advance cancer treatment. Here, we review literature focused on the molecular control of DCs, with a particular emphasis on cytokine- and STAT-mediated DC regulation. In addition, we highlight recent studies that delineate the importance of DCs in anti-tumor immunity and immune therapy, with the overall goal of improving knowledge of tumor-associated factors and intrinsic DC signaling cascades that influence DC function in cancer.
Collapse
Affiliation(s)
- Taylor T Chrisikos
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Yifan Zhou
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Natalie Slone
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rachel Babcock
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Stephanie S Watowich
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, 77030, USA.
| | - Haiyan S Li
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
21
|
A Characterization of Dendritic Cells and Their Role in Immunotherapy in Glioblastoma: From Preclinical Studies to Clinical Trials. Cancers (Basel) 2019; 11:cancers11040537. [PMID: 30991681 PMCID: PMC6521200 DOI: 10.3390/cancers11040537] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma (GBM) is the most common and fatal primary central nervous system malignancy in adults with a median survival of less than 15 months. Surgery, radiation, and chemotherapy are the standard of care and provide modest benefits in survival, but tumor recurrence is inevitable. The poor prognosis of GBM has made the development of novel therapies targeting GBM of paramount importance. Immunotherapy via dendritic cells (DCs) has garnered attention and research as a potential strategy to boost anti-tumor immunity in recent years. As the “professional” antigen processing and presenting cells, DCs play a key role in the initiation of anti-tumor immune responses. Pre-clinical studies in GBM have shown long-term tumor survival and immunological memory in murine models with stimulation of DC activity with various antigens and costimulatory molecules. Phase I and II clinical trials of DC vaccines in GBM have demonstrated some efficacy in improving the median overall survival with minimal to no toxicity with promising initial results from the first Phase III trial. However, there remains no standardization of vaccines in terms of which antigens are used to pulse DCs ex vivo, sites of DC injection, and optimal adjuvant therapies. Future work with DC vaccines aims to elucidate the efficacy of DC-based therapy alone or in combination with other immunotherapy adjuvants in additional Phase III trials.
Collapse
|
22
|
Christiansen AJ, Dieterich LC, Ohs I, Bachmann SB, Bianchi R, Proulx ST, Hollmén M, Aebischer D, Detmar M. Lymphatic endothelial cells attenuate inflammation via suppression of dendritic cell maturation. Oncotarget 2018; 7:39421-39435. [PMID: 27270646 PMCID: PMC5129942 DOI: 10.18632/oncotarget.9820] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 05/25/2016] [Indexed: 12/26/2022] Open
Abstract
Vascular endothelial growth factor-C (VEGF-C)-induced lymphangiogenesis and increased tissue drainage have been reported to inhibit acute and chronic inflammation, and an activated lymphatic endothelium might mediate peripheral tolerance. Using transgenic mice overexpressing VEGF-C in the skin, we found that under inflammatory conditions, VEGF-C-mediated expansion of the cutaneous lymphatic network establishes an immune-inhibitory microenvironment characterised by increased regulatory T (Treg) cells, immature CD11c+CD11b+ dendritic cells (DCs) and CD8+ cells exhibiting decreased effector function. Strikingly, lymphatic endothelial cell (LEC)-conditioned media (CM) potently suppress DC maturation with reduced expression of MHCII, CD40, and IL-6, and increased IL-10 and CCL2 expression. We identify an imbalance in prostaglandin synthase expression after LEC activation, favoring anti-inflammatory prostacyclin synthesis. Importantly, blockade of LEC prostaglandin synthesis partially restores DC maturity. LECs also produce TGF-ß1, contributing to the immune-inhibitory microenvironment. This study identifies novel mechanisms by which the lymphatic endothelium modulates cellular immune responses to limit inflammation.
Collapse
Affiliation(s)
- Ailsa J Christiansen
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich, Switzerland
| | - Lothar C Dieterich
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich, Switzerland
| | - Isabel Ohs
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich, Switzerland
| | - Samia B Bachmann
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich, Switzerland
| | - Roberta Bianchi
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich, Switzerland
| | - Steven T Proulx
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich, Switzerland
| | - Maija Hollmén
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich, Switzerland
| | - David Aebischer
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich, Switzerland
| | - Michael Detmar
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
23
|
Theron AJ, Anderson R, Rossouw TM, Steel HC. The Role of Transforming Growth Factor Beta-1 in the Progression of HIV/AIDS and Development of Non-AIDS-Defining Fibrotic Disorders. Front Immunol 2017; 8:1461. [PMID: 29163528 PMCID: PMC5673850 DOI: 10.3389/fimmu.2017.01461] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/18/2017] [Indexed: 12/21/2022] Open
Abstract
Even after attainment of sustained viral suppression following implementation of highly active antiretroviral therapy, HIV-infected persons continue to experience persistent, low-grade, systemic inflammation. Among other mechanisms, this appears to result from ongoing microbial translocation from a damaged gastrointestinal tract. This HIV-related chronic inflammatory response is paralleled by counteracting, but only partially effective, biological anti-inflammatory processes. Paradoxically, however, this anti-inflammatory response not only exacerbates immunosuppression but also predisposes for development of non-AIDS-related, non-communicable disorders. With respect to the pathogenesis of both sustained immunosuppression and the increased frequency of non-AIDS-related disorders, the anti-inflammatory/profibrotic cytokine, transforming growth factor-β1 (TGF-β1), which remains persistently elevated in both untreated and virally suppressed HIV-infected persons, may provide a common link. In this context, the current review is focused on two different, albeit related, harmful activities of TGF-β1 in HIV infection. First, on the spectrum of anti-inflammatory/immunosuppressive activities of TGF-β1 and the involvement of this cytokine, derived predominantly from T regulatory cells, in driving disease progression in HIV-infected persons via both non-fibrotic and profibrotic mechanisms. Second, the possible involvement of sustained elevations in circulating and tissue TGF-β1 in the pathogenesis of non-AIDS-defining cardiovascular, hepatic, pulmonary and renal disorders, together with a brief comment on potential TGF-β1-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Annette J. Theron
- Faculty of Health Sciences, Department of Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
- Tshwane Academic Division of the National Health Laboratory Service, Pretoria, South Africa
| | - Ronald Anderson
- Faculty of Health Sciences, Department of Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
| | - Theresa M. Rossouw
- Faculty of Health Sciences, Department of Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
| | - Helen C. Steel
- Faculty of Health Sciences, Department of Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
24
|
Abstract
Transforming growth factor βs (TGF-βs) are closely related ligands that have pleiotropic activity on most cell types of the body. They act through common heterotetrameric TGF-β type II and type I transmembrane dual specificity kinase receptor complexes, and the outcome of signaling is context-dependent. In normal tissue, they serve a role in maintaining homeostasis. In many diseased states, particularly fibrosis and cancer, TGF-β ligands are overexpressed and the outcome of signaling is diverted toward disease progression. There has therefore been a concerted effort to develop drugs that block TGF-β signaling for therapeutic benefit. This review will cover the basics of TGF-β signaling and its biological activities relevant to oncology, present a summary of pharmacological TGF-β blockade strategies, and give an update on preclinical and clinical trials for TGF-β blockade in a variety of solid tumor types.
Collapse
Affiliation(s)
- Rosemary J Akhurst
- Department of Anatomy and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California 94158-9001
| |
Collapse
|
25
|
Kim SY, Kang D, Choi HJ, Joo Y, Kim JH, Song JJ. Prime-boost immunization by both DNA vaccine and oncolytic adenovirus expressing GM-CSF and shRNA of TGF-β2 induces anti-tumor immune activation. Oncotarget 2017; 8:15858-15877. [PMID: 28178658 PMCID: PMC5362529 DOI: 10.18632/oncotarget.15008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 12/31/2016] [Indexed: 12/21/2022] Open
Abstract
A successful DNA vaccine for the treatment of tumors should break established immune tolerance to tumor antigen. However, due to the relatively low immunogenicity of DNA vaccines, compared to other kinds of vaccines using live virus or protein, a recombinant viral vector was used to enhance humoral and cellular immunity. In the current study, we sought to develop a novel anti-cancer agent as a complex of DNA and oncolytic adenovirus for the treatment of malignant melanoma in the C57BL/6 mouse model. MART1, a human melanoma-specific tumor antigen, was used to induce an increased immune reaction, since a MART1-protective response is required to overcome immune tolerance to the melanoma antigen MelanA. Because GM-CSF is a potent inducer of anti-tumor immunity and TGF-β2 is involved in tumor survival and host immune suppression, mouse GM-CSF (mGM-CSF) and shRNA of mouse TGF-β2 (shmTGF-β2) genes were delivered together with MART1 via oncolytic adenovirus. MART1 plasmid was also used for antigen-priming. To compare the anti-tumor effect of oncolytic adenovirus expressing both mGM-CSF and shmTGF-β2 (AdGshT) with that of oncolytic adenovirus expressing mGM-CSF only (AdG), each virus was intratumorally injected into melanoma-bearing C57BL/6 mice. As a result, mice that received AdGshT showed delayed tumor growth than those that received AdG. Heterologous prime-boost immunization was combined with oncolytic AdGshT and MART1 expression to result in further delayed tumor growth. This regression is likely due to the following 4 combinations: MART1-derived mouse melanoma antigen-specific immune reaction, immune stimulation by mGM-CSF/shmTGF-β2, tumor growth inhibition by shmTGF-β2, and tumor cell-specific lysis via an oncolytic adenovirus. Immune activation was mainly induced by mature tumor-infiltrating dendritic cell (TIDC) and lowered regulatory T cells in tumor-infiltrating lymphocytes (TIL). Taken together, these findings demonstrate that human MART1 induces a mouse melanoma antigen-specific immune reaction. In addition, the results also indicate that combination therapy of MART1 plasmid, together with an oncolytic adenovirus expressing MART1, mGM-CSF, and shmTGF-β2, is a promising candidate for the treatment of malignant melanoma.
Collapse
Affiliation(s)
- So Young Kim
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Korea
| | - Dongxu Kang
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Korea.,Department of Oncology, Affiliated Hospital of Yanbian University, Yanji, Jilin Province, P.R. China
| | - Hye Jin Choi
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Yeonsoo Joo
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Joo-Hang Kim
- CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Jae J Song
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
26
|
Chakraborty K, Chatterjee S, Bhattacharyya A. Modulation of CD11c+ lung dendritic cells in respect to TGF-β in experimental pulmonary fibrosis. Cell Biol Int 2017; 41:991-1000. [PMID: 28557137 DOI: 10.1002/cbin.10800] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/25/2017] [Indexed: 12/30/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a deadly, progressive lung disease with very few treatment options till now. Bleomycin-induced pulmonary fibrosis (BIPF) is a commonly used mice model in IPF research. TGF-β1 has been shown to play a key role in pulmonary fibrosis (PF). Dendritic cell (DC) acts as a bridge between innate and adaptive immune systems. The coexistence of chronic inflammation sustained by mature DCs with fibrosis suggests that inflammatory phenomenon has key importance in the pathogenesis of pulmonary fibrosis. Here, we investigated the modulation of DCs phenotypic maturation, accumulation in lung tissue, and expression of other lung DC subsets in respect to TGF-β in PF. First, we established BIPF model in mice and blocked TGF-β expression by the use of inhibitor SB431542. Accumulation of lung CD11c+ DCs is significantly higher in both inflammatory and fibrotic phases of the disease but that percentages got reduced in the absence of TGF-β. TGF-β initiates up-regulation of costimulatory molecules CD86 and CD80 in the inflammatory phases of the disease but not so at fibrotic stage. Expression of lung DC subset CD11c+CD103+ is significantly increased in inflammatory phase and also in fibrotic phase of BIPF. Blocking of TGF-β causes decreased expression of CD11c+CD103+ DCs. Another important lung DC subset CD11c+CD11b+ expression is suppressed by the absence of TGF-β after bleomycin administration. CD11c+CD103+ DCs might have anti-inflammatory as well as anti-fibrotic nature in PF. All these data demonstrate differential modulation of CD11c+ lung DCs by TGF-β in experimental PF.
Collapse
Affiliation(s)
- Kaustav Chakraborty
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Soumya Chatterjee
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Arindam Bhattacharyya
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| |
Collapse
|
27
|
Wang H, Franco F, Ho PC. Metabolic Regulation of Tregs in Cancer: Opportunities for Immunotherapy. Trends Cancer 2017; 3:583-592. [PMID: 28780935 DOI: 10.1016/j.trecan.2017.06.005] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/18/2017] [Accepted: 06/20/2017] [Indexed: 02/06/2023]
Abstract
The promising outcomes observed in cancer immunotherapy are evidence that the immune system provides a powerful arsenal for the restriction of tumor outgrowth; however, the immunosuppressive tumor microenvironment (TME) is known to impair antitumor immunity and impede the efficacy of cancer immunotherapies. Regulatory T cells (Tregs), which prevent overt immune responses and autoimmunity, accumulate aberrantly in some types of tumor to suppress antitumor immunity and support the establishment of an immunosuppressive microenvironment. Ablation of Tregs has been shown to not only unleash antitumor immunity and interrupt formation of an immunosuppressive TME, but also lead to severe autoimmune disorders. Therefore, it is essential to develop approaches to specifically target intratumoral Tregs. Herein, we summarize the immunomodulatory functions of Tregs in the TME and discuss how metabolic regulation of Tregs can facilitate intratumoral Treg accumulation.
Collapse
Affiliation(s)
- Haiping Wang
- Department of Fundamental Oncology, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Vaud, Switzerland; Ludwig Lausanne Branch, Epalinges, Vaud, Switzerland
| | - Fabien Franco
- Department of Fundamental Oncology, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Vaud, Switzerland; Ludwig Lausanne Branch, Epalinges, Vaud, Switzerland
| | - Ping-Chih Ho
- Department of Fundamental Oncology, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Vaud, Switzerland; Ludwig Lausanne Branch, Epalinges, Vaud, Switzerland.
| |
Collapse
|
28
|
Sanjabi S, Oh SA, Li MO. Regulation of the Immune Response by TGF-β: From Conception to Autoimmunity and Infection. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022236. [PMID: 28108486 DOI: 10.1101/cshperspect.a022236] [Citation(s) in RCA: 410] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transforming growth factor β (TGF-β) is a pleiotropic cytokine involved in both suppressive and inflammatory immune responses. After 30 years of intense study, we have only begun to elucidate how TGF-β alters immunity under various conditions. Under steady-state conditions, TGF-β regulates thymic T-cell selection and maintains homeostasis of the naïve T-cell pool. TGF-β inhibits cytotoxic T lymphocyte (CTL), Th1-, and Th2-cell differentiation while promoting peripheral (p)Treg-, Th17-, Th9-, and Tfh-cell generation, and T-cell tissue residence in response to immune challenges. Similarly, TGF-β controls the proliferation, survival, activation, and differentiation of B cells, as well as the development and functions of innate cells, including natural killer (NK) cells, macrophages, dendritic cells, and granulocytes. Collectively, TGF-β plays a pivotal role in maintaining peripheral tolerance against self- and innocuous antigens, such as food, commensal bacteria, and fetal alloantigens, and in controlling immune responses to pathogens.
Collapse
Affiliation(s)
- Shomyseh Sanjabi
- Institute of Virology and Immunology, Gladstone Institutes, San Francisco, California 94158.,Department of Microbiology and Immunology, University of California, San Francisco, California 94143
| | - Soyoung A Oh
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Ming O Li
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| |
Collapse
|
29
|
Lerrer S, Liubomirski Y, Bott A, Abnaof K, Oren N, Yousaf A, Körner C, Meshel T, Wiemann S, Ben-Baruch A. Co-Inflammatory Roles of TGFβ1 in the Presence of TNFα Drive a Pro-inflammatory Fate in Mesenchymal Stem Cells. Front Immunol 2017; 8:479. [PMID: 28553282 PMCID: PMC5425596 DOI: 10.3389/fimmu.2017.00479] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 04/05/2017] [Indexed: 12/19/2022] Open
Abstract
High plasticity is a hallmark of mesenchymal stem cells (MSCs), and as such, their differentiation and activities may be shaped by factors of their microenvironment. Bones, tumors, and cardiomyopathy are examples of niches and conditions that contain MSCs and are enriched with tumor necrosis factor α (TNFα) and transforming growth factor β1 (TGFβ1). These two cytokines are generally considered as having opposing roles in regulating immunity and inflammation (pro- and anti-inflammatory, respectively). Here, we performed global gene expression analysis of human bone marrow-derived MSCs and identified overlap in half of the transcriptional programs that were modified by TNFα and TGFβ1. The two cytokines elevated the mRNA expression of soluble factors, including mRNAs of pro-inflammatory mediators. Accordingly, the typical pro-inflammatory factor TNFα prominently induced the protein expression levels of the pro-inflammatory mediators CCL2, CXCL8 (IL-8), and cyclooxygenase-2 (Cox-2) in MSCs, through the NF-κB/p65 pathway. In parallel, TGFβ1 did not elevate CXCL8 protein levels and induced the protein expression of CCL2 at much lower levels than TNFα; yet, TGFβ1 readily induced Cox-2 and acted predominantly via the Smad3 pathway. Interestingly, combined stimulation of MSCs by TNFα + TGFβ1 led to a cooperative induction of all three inflammatory mediators, indicating that TGFβ1 functioned as a co-inflammatory cytokine in the presence of TNFα. The cooperative activities of TNFα + TGFβ1 that have led to CCL2 and CXCL8 induction were almost exclusively dependent on p65 activation and were not regulated by Smad3 or by the upstream regulator TGFβ-activated kinase 1 (TAK1). In contrast, the TNFα + TGFβ1-induced cooperative elevation in Cox-2 was mostly dependent on Smad3 (demonstrating cooperativity with activated NF-κB) and was partly regulated by TAK1. Studies with MSCs activated by TNFα + TGFβ1 revealed that they release factors that can affect other cells in their microenvironment and induce breast tumor cell elongation, migration, and scattering out of spheroid tumor masses. Thus, our findings demonstrate a TNFα + TGFβ1-driven pro-inflammatory fate in MSCs, identify specific molecular mechanisms involved, and propose that TNFα + TGFβ1-stimulated MSCs influence the tumor niche. These observations suggest key roles for the microenvironment in regulating MSC functions, which in turn may affect different health-related conditions.
Collapse
Affiliation(s)
- Shalom Lerrer
- Faculty of Life Sciences, Department of Cell Research and Immunology, Tel Aviv University, Tel Aviv, Israel
| | - Yulia Liubomirski
- Faculty of Life Sciences, Department of Cell Research and Immunology, Tel Aviv University, Tel Aviv, Israel
| | - Alexander Bott
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Khalid Abnaof
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nino Oren
- Faculty of Life Sciences, Department of Cell Research and Immunology, Tel Aviv University, Tel Aviv, Israel
| | - Afsheen Yousaf
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cindy Körner
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tsipi Meshel
- Faculty of Life Sciences, Department of Cell Research and Immunology, Tel Aviv University, Tel Aviv, Israel
| | - Stefan Wiemann
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Adit Ben-Baruch
- Faculty of Life Sciences, Department of Cell Research and Immunology, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
30
|
Klein-Szanto AJ, Bassi DE. Proprotein convertase inhibition: Paralyzing the cell's master switches. Biochem Pharmacol 2017; 140:8-15. [PMID: 28456517 DOI: 10.1016/j.bcp.2017.04.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 04/25/2017] [Indexed: 12/18/2022]
Abstract
Proprotein convertases are serine proteases responsible for the cleavage and subsequent activation of protein substrates, many of them relevant for the development of an ample variety of diseases. Seven of the PCs, including furin and PACE4, recognize and hydrolyze the C-terminal end of the general sequence RXRR/KXR, whereas PCSK-9 recognizes a series of non-basic amino acids. In some systems, PC-mediated substrate activation results in the development of pathological processes, such as cancer, endocrinopathies, and cardiovascular and infectious diseases. After establishing PCs as relevant contributors to disease processes, research efforts were directed towards the development of inhibition strategies, including small and large molecules, anti-sense therapies, and antibody-based therapies. Most of these inhibitors mimic the consensus sequence of PCs, blocking the active site in a competitive manner. The most promising inhibitors were designed as bioengineered proteins; however, some non-protein and peptidomimetic agents have also proved to be effective. These efforts led to the design of pre-clinical studies and clinical trials utilizing inhibitors to PCs. Although the initial studies were performed using non-selective PCs inhibitors, such as CMK, the search for more specific, and compartmentalized selective inhibitors resulted in specific activities ascribed to some, but not all of the PCs. For instance, PACE4 inhibitors were effective in decreasing prostate cancer cell proliferation, and neovascularization. Decreased metastatic ovarian cancer utilizing furin inhibitors represents one of the major endeavors, currently in a phase II trial stage. Antibodies targeting PCSK-9 decreased significantly the levels of HDL-cholesterol, in a phase III trial. The study of Proprotein convertases has reached a stage of maturity. New strategies based on the alteration of their activity at the cellular and clinical level represent a promising experimental pharmacology field. The development of allosteric inhibitors, or specific agents directed against individual PCs is one of the challenges to be unraveled in the future.
Collapse
Affiliation(s)
| | - Daniel E Bassi
- Fox Chase Cancer Center, 333 Cotman Ave, Philadelphia 19111, USA.
| |
Collapse
|
31
|
Abdollahi E, Momtazi AA, Johnston TP, Sahebkar A. Therapeutic effects of curcumin in inflammatory and immune‐mediated diseases: A nature‐made jack‐of‐all‐trades? J Cell Physiol 2017; 233:830-848. [DOI: 10.1002/jcp.25778] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 01/05/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Elham Abdollahi
- Department of Medical ImmunologySchool of Medicine, Mashhad University of Medical SciencesMashhadIran
- Student Research CommitteeMashhad University of Medical SciencesMashhadIran
| | - Amir Abbas Momtazi
- Student Research Committee, Nanotechnology Research Center, Department of Medical BiotechnologySchool of Medicine, Mashhad University of Medical SciencesMashhadIran
| | - Thomas P. Johnston
- Division of Pharmaceutical SciencesSchool of Pharmacy, University of Missouri‐Kansas CityKansas CityMissouri
| | - Amirhossein Sahebkar
- Biotechnology Research CenterMashhad University of Medical SciencesMashhadIran
- Neurogenic Inflammation Research CenterMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
32
|
Abstract
TGF-β is an anti-inflammatory cytokine whose signaling is negatively controlled by Smad7. Previously, we established a role for Smad7 in the generation of autoreactive T cells; however, the function of Smad7 in dendritic cells (DCs) remains elusive. Here, we demonstrate that DC-specific Smad7 deficiency resulted in elevated expression of the transcription factors Batf3 and IRF8, leading to increased frequencies of CD8+CD103+ DCs in the spleen. Furthermore, Smad7-deficient DCs expressed higher levels of indoleamine 2,3-dioxygenase (IDO), an enzyme associated with tolerance induction. Mice devoid of Smad7 specifically in DCs are resistant to the development of experimental autoimmune encephalomyelitis (EAE) as a result of an increase of protective regulatory T cells (Tregs) and reduction of encephalitogenic effector T cells in the central nervous system. In agreement, inhibition of IDO activity or depletion of Tregs restored disease susceptibility. Intriguingly, when Smad7-deficient DCs also lacked the IFN-γ receptor, the mice regained susceptibility to EAE, demonstrating that IFN-γ signaling in DCs mediates their tolerogenic function. Our data indicate that Smad7 expression governs splenic DC subset differentiation and is critical for the promotion of their efficient function in immunity.
Collapse
|
33
|
Cantelli G, Crosas-Molist E, Georgouli M, Sanz-Moreno V. TGFΒ-induced transcription in cancer. Semin Cancer Biol 2017; 42:60-69. [PMID: 27586372 PMCID: PMC6137079 DOI: 10.1016/j.semcancer.2016.08.009] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/19/2016] [Indexed: 12/15/2022]
Abstract
The Transforming Growth Factor-beta (TGFβ) pathway mediates a broad spectrum of cellular processes and is involved in several diseases, including cancer. TGFβ has a dual role in tumours, acting as a tumour suppressor in the early phase of tumorigenesis and as a tumour promoter in more advanced stages. In this review, we discuss the effects of TGFβ-driven transcription on all stages of tumour progression, with special focus on lung cancer. Since some TGFβ target genes are specifically involved in promoting metastasis, we speculate that these genes might be good targets to block tumour progression without compromising the tumour suppressor effects of the TGFβ pathway.
Collapse
Affiliation(s)
- Gaia Cantelli
- Tumour Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Eva Crosas-Molist
- Tumour Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Mirella Georgouli
- Tumour Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Victoria Sanz-Moreno
- Tumour Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK.
| |
Collapse
|
34
|
Oh J, Barve M, Matthews CM, Koon EC, Heffernan TP, Fine B, Grosen E, Bergman MK, Fleming EL, DeMars LR, West L, Spitz DL, Goodman H, Hancock KC, Wallraven G, Kumar P, Bognar E, Manning L, Pappen BO, Adams N, Senzer N, Nemunaitis J. Phase II study of Vigil® DNA engineered immunotherapy as maintenance in advanced stage ovarian cancer. Gynecol Oncol 2016; 143:504-510. [DOI: 10.1016/j.ygyno.2016.09.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/15/2016] [Accepted: 09/19/2016] [Indexed: 12/19/2022]
|
35
|
Xue D, Liang Y, Duan S, He J, Su J, Zhu J, Hu N, Liu J, Zhao Y, Lu X. Enhanced anti-tumor immunity against breast cancer induced by whole tumor cell vaccines genetically modified expressing α-Gal epitopes. Oncol Rep 2016; 36:2843-2851. [PMID: 27666541 DOI: 10.3892/or.2016.5128] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 06/27/2016] [Indexed: 11/05/2022] Open
Abstract
Whole tumor cell vaccines have shown much promise, but demonstrated poor efficiency in phase III trials. In this study, we modified MDA-MB‑231 tumor cells (MDA-MB‑231Gal+) to express α-1, 3-galactosyltransferase (α-1, 3-GT) protein, to potentially enhance antitumor effect of whole tumor cell vaccines. MDA-MB‑231 tumor cell vaccines were transfected with a reconstructed lentiviral containing α-1, 3-GT genes. Tumor growth, tumorigenesis and survival of Hu-NOD-SCID mice were observed when tumor-bearing mice were injected with tumor cell vaccines. Proliferation and apoptosis in MDA-MB‑231 tumor xenografts were observed by immunohistochemistry. The levels of cytokine secretion in the serum of mice were tested by ELISA. CD8+ T cells infiltrating tumors were assessed by flow cytometry. MDA-MB‑231Gal+ cells expressed active α-1, 3-GT and produced α-Gal in vitro. MDA-MB‑231Gal+ cell vaccines suppressed tumor growth and tumorigenesis in immunized Hu-NOD-SCID mice. Additionally, decrease of TGF-β, IL-10 and increase of INF-γ, IL-12 were observed in tumor cell vaccinated mice. Furthermore, the cell vaccines enhanced infiltration of cytotoxic CD8+ T cells in the tumor microenvironment of immunized mice. The MDA-MB‑231Gal+ cell vaccines modified α-1, 3-GT genes improved the antitumor effect.
Collapse
Affiliation(s)
- Dabing Xue
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Ying Liang
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Siliang Duan
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jian He
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jing Su
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jianmeng Zhu
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Nan Hu
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jianming Liu
- Department of Respiratoy Diseases, The Third Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Yongxiang Zhao
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiaoling Lu
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
36
|
Dysregulation of TGFβ1 Activity in Cancer and Its Influence on the Quality of Anti-Tumor Immunity. J Clin Med 2016; 5:jcm5090076. [PMID: 27589814 PMCID: PMC5039479 DOI: 10.3390/jcm5090076] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 08/26/2016] [Accepted: 08/29/2016] [Indexed: 01/01/2023] Open
Abstract
TGFβ1 is a pleiotropic cytokine that exhibits a variety of physiologic and immune regulatory functions. Although its influence on multiple cell types is critical for the regulation of numerous biologic processes in the host, dysregulation of both TGFβ1 expression and activity is frequently observed in cancer and contributes to various aspects of cancer progression. This review focuses on TGFβ1’s contribution to tumor immune suppression and escape, with emphasis on the influence of this regulatory cytokine on the differentiation and function of dendritic cells and T cells. Clinical trials targeting TGFβ1 in cancer patients are also reviewed, and strategies for future therapeutic interventions that build on our current understanding of immune regulation by TGFβ1 are discussed.
Collapse
|
37
|
Lasigliè D, Boero S, Bauer I, Morando S, Damonte P, Cea M, Monacelli F, Odetti P, Ballestrero A, Uccelli A, Mostoslavsky R, Poggi A, Nencioni A. Sirt6 regulates dendritic cell differentiation, maturation, and function. Aging (Albany NY) 2016; 8:34-49. [PMID: 26761436 PMCID: PMC4761712 DOI: 10.18632/aging.100870] [Citation(s) in RCA: 25] [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: 11/12/2015] [Accepted: 12/29/2015] [Indexed: 12/29/2022]
Abstract
Dendritic cells (DCs) are antigen-presenting cells that critically influence decisions about immune activation or tolerance. Impaired DC function is at the core of common chronic disorders and contributes to reduce immunocompetence during aging. Knowledge on the mechanisms regulating DC generation and function is necessary to understand the immune system and to prevent disease and immunosenescence. Here we show that the sirtuin Sirt6, which was previously linked to healthspan promotion, stimulates the development of myeloid, conventional DCs (cDCs). Sirt6-knockout (Sirt6KO) mice exhibit low frequencies of bone marrow cDC precursors and low yields of bone marrow-derived cDCs compared to wild-type (WT) animals. Sirt6KO cDCs express lower levels of class II MHC, of costimulatory molecules, and of the chemokine receptor CCR7, and are less immunostimulatory compared to WT cDCs. Similar effects in terms of differentiation and immunostimulatory capacity were observed in human monocyte-derived DCs in response to SIRT6 inhibition. Finally, while Sirt6KO cDCs show an overall reduction in their ability to produce IL-12, TNF-α and IL-6 secretion varies dependent on the stimulus, being reduced in response to CpG, but increased in response to other Toll-like receptor ligands. In conclusion, Sirt6 plays a crucial role in cDC differentiation and function and reduced Sirt6 activity may contribute to immunosenescence.
Collapse
Affiliation(s)
- Denise Lasigliè
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
| | - Silvia Boero
- The Istituto di Ricovero e Cura a Carattere Scientifico Azienda Ospedaliera Universitaria San Martino-Istituto Scientifico Tumori, Istituto Nazionale per la Ricerca sul Cancro, 16132 Genoa, Italy
| | - Inga Bauer
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
| | - Sara Morando
- Department of Neurology, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16100 Genoa, Italy
| | - Patrizia Damonte
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
| | - Michele Cea
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
| | - Fiammetta Monacelli
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- The Istituto di Ricovero e Cura a Carattere Scientifico Azienda Ospedaliera Universitaria San Martino-Istituto Scientifico Tumori, Istituto Nazionale per la Ricerca sul Cancro, 16132 Genoa, Italy
| | - Paizio Odetti
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- The Istituto di Ricovero e Cura a Carattere Scientifico Azienda Ospedaliera Universitaria San Martino-Istituto Scientifico Tumori, Istituto Nazionale per la Ricerca sul Cancro, 16132 Genoa, Italy
| | - Alberto Ballestrero
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- The Istituto di Ricovero e Cura a Carattere Scientifico Azienda Ospedaliera Universitaria San Martino-Istituto Scientifico Tumori, Istituto Nazionale per la Ricerca sul Cancro, 16132 Genoa, Italy
| | - Antonio Uccelli
- The Istituto di Ricovero e Cura a Carattere Scientifico Azienda Ospedaliera Universitaria San Martino-Istituto Scientifico Tumori, Istituto Nazionale per la Ricerca sul Cancro, 16132 Genoa, Italy
- Department of Neurology, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16100 Genoa, Italy
| | - Raul Mostoslavsky
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114 USA
| | - Alessandro Poggi
- The Istituto di Ricovero e Cura a Carattere Scientifico Azienda Ospedaliera Universitaria San Martino-Istituto Scientifico Tumori, Istituto Nazionale per la Ricerca sul Cancro, 16132 Genoa, Italy
| | - Alessio Nencioni
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- The Istituto di Ricovero e Cura a Carattere Scientifico Azienda Ospedaliera Universitaria San Martino-Istituto Scientifico Tumori, Istituto Nazionale per la Ricerca sul Cancro, 16132 Genoa, Italy
| |
Collapse
|
38
|
Ludewig P, Gallizioli M, Urra X, Behr S, Brait VH, Gelderblom M, Magnus T, Planas AM. Dendritic cells in brain diseases. Biochim Biophys Acta Mol Basis Dis 2015; 1862:352-67. [PMID: 26569432 DOI: 10.1016/j.bbadis.2015.11.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 11/05/2015] [Accepted: 11/05/2015] [Indexed: 12/25/2022]
Affiliation(s)
- Peter Ludewig
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mattia Gallizioli
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | - Xabier Urra
- Functional Unit of Cerebrovascular Diseases, Hospital Clínic, Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Sarah Behr
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa H Brait
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna M Planas
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
39
|
Melanoma-derived factors alter the maturation and activation of differentiated tissue-resident dendritic cells. Immunol Cell Biol 2015; 94:24-38. [PMID: 26010746 DOI: 10.1038/icb.2015.58] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 05/05/2015] [Accepted: 05/20/2015] [Indexed: 01/26/2023]
Abstract
Dendritic cells (DCs) are key regulators of host immunity that are capable of inducing either immune tolerance or activation. In addition to their well-characterized role in shaping immune responses to foreign pathogens, DCs are also known to be critical for the induction and maintenance of anti-tumor immune responses. Therefore, it is important to understand how tumors influence the function of DCs and the quality of immune responses they elicit. Although the majority of studies in this field to date have utilized either immortalized DC lines or DC populations that have been generated under artificial conditions from hematopoietic precursors in vitro, we wished to investigate how tumors impact the function of already differentiated, tissue-resident DCs. Therefore, we used both an ex vivo and in vivo model system to assess the influence of melanoma-derived factors on DC maturation and activation. In ex vivo studies with freshly isolated splenic DCs, we demonstrate that the extent to which DC maturation and activation are altered by these factors correlates with melanoma tumorigenicity, and we identify partial roles for tumor-derived transforming growth factor (TGF)β1 and vascular endothelial growth factor (VEGF)-A in the altered functionality of DCs. In vivo studies using a lung metastasis model of melanoma also demonstrate tumorigenicity-dependent alterations to the function of lung-resident DCs, and skewed production of proinflammatory cytokines and chemokines by these tumor-altered cells is associated with recruitment of an immune infiltrate that may ultimately favor tumor immune escape and outgrowth.
Collapse
|
40
|
Whole Tumor Antigen Vaccines: Where Are We? Vaccines (Basel) 2015; 3:344-72. [PMID: 26343191 PMCID: PMC4494356 DOI: 10.3390/vaccines3020344] [Citation(s) in RCA: 198] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 04/13/2015] [Accepted: 04/16/2015] [Indexed: 12/19/2022] Open
Abstract
With its vast amount of uncharacterized and characterized T cell epitopes available for activating CD4+ T helper and CD8+ cytotoxic lymphocytes simultaneously, whole tumor antigen represents an attractive alternative source of antigens as compared to tumor-derived peptides and full-length recombinant tumor proteins for dendritic cell (DC)-based immunotherapy. Unlike defined tumor-derived peptides and proteins, whole tumor lysate therapy is applicable to all patients regardless of their HLA type. DCs are essentially the master regulators of immune response, and are the most potent antigen-presenting cell population for priming and activating naïve T cells to target tumors. Because of these unique properties, numerous DC-based immunotherapies have been initiated in the clinics. In this review, we describe the different types of whole tumor antigens that we could use to pulse DCs ex vivo and in vivo. We also discuss the different routes of delivering whole tumor antigens to DCs in vivo and activating them with toll-like receptor agonists.
Collapse
|
41
|
Contreras-Ruiz L, Masli S. Immunomodulatory cross-talk between conjunctival goblet cells and dendritic cells. PLoS One 2015; 10:e0120284. [PMID: 25793763 PMCID: PMC4368435 DOI: 10.1371/journal.pone.0120284] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 01/27/2015] [Indexed: 12/03/2022] Open
Abstract
Goblet cells are secretory epithelial cells of mucosal tissues that confer protection from environmental agents or pathogens via expression and secretion of soluble mucins. Loss of these cells is associated with several chronic inflammatory disorders of the mucosa. Although demonstrated to transfer antigens from the luminal surface to stromal cells in the intestinal mucosa, it is not known if goblet cells contribute to the regulation of an immune response. In this study we report that similar to intestinal and respiratory mucosal epithelia, mouse ocular surface epithelia predominantly express the TGF-ß2 isoform. Specifically, we demonstrate the ability of goblet cells to express TGF-ß2 and increase it in response to Toll-Like Receptor 4 mediated stimulus in cultures. Goblet cells not only express TGF-ß2, but are also able to activate it in a thrombospondin-1 (TSP-1) dependent manner via their cell surface receptor CD36. Furthermore, goblet cell derived soluble factors that possibly include TGF-ß2, alter dendritic cell (DC) phenotype to a tolerogenic type by downregulating DC expression of MHC class II and co-stimulatory molecules CD80, CD86 and CD40. Thus our study demonstrates goblet cells as a cellular source of active TGF-ß2 in ocular mucosa and implicates their immunomodulatory function in maintaining mucosal immune homeostasis.
Collapse
Affiliation(s)
- Laura Contreras-Ruiz
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Sharmila Masli
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
42
|
Sulforaphane epigenetically regulates innate immune responses of porcine monocyte-derived dendritic cells induced with lipopolysaccharide. PLoS One 2015; 10:e0121574. [PMID: 25793534 PMCID: PMC4368608 DOI: 10.1371/journal.pone.0121574] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 02/17/2015] [Indexed: 12/22/2022] Open
Abstract
Histone acetylation, regulated by histone deacetylases (HDACs) is a key epigenetic mechanism controlling gene expressions. Although dendritic cells (DCs) are playing pivotal roles in host immune responses, the effect of epigenetic modulation of DCs immune responses remains unknown. Sulforaphane (SFN) as a HDAC inhibitor has anti-inflammatory properties, which is used to investigate the epigenetic regulation of LPS-induced immune gene and HDAC family gene expressions in porcine monocyte-derived dendritic cells (moDCs). SFN was found to inhibit the lipopolysaccharide LPS induced HDAC6, HDAC10 and DNA methyltransferase (DNMT3a) gene expression, whereas up-regulated the expression of DNMT1 gene. Additionally, SFN was observed to inhibit the global HDAC activity, and suppressed moDCs differentiation from immature to mature DCs through down-regulating the CD40, CD80 and CD86 expression and led further to enhanced phagocytosis of moDCs. The SFN pre-treated of moDCs directly altered the LPS-induced TLR4 and MD2 gene expression and dynamically regulated the TLR4-induced activity of transcription factor NF-κB and TBP. SFN showed a protective role in LPS induced cell apoptosis through suppressing the IRF6 and TGF-ß1 production. SFN impaired the pro-inflammatory cytokine TNF-α and IL-1ß secretion into the cell culture supernatants that were induced in moDCs by LPS stimulation, whereas SFN increased the cellular-resident TNF-α accumulation. This study demonstrates that through the epigenetic mechanism the HDAC inhibitor SFN could modulate the LPS induced innate immune responses of porcine moDCs.
Collapse
|
43
|
Nayar S, Dasgupta P, Galustian C. Extending the lifespan and efficacies of immune cells used in adoptive transfer for cancer immunotherapies-A review. Oncoimmunology 2015; 4:e1002720. [PMID: 26155387 DOI: 10.1080/2162402x.2014.1002720] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/19/2014] [Accepted: 12/20/2014] [Indexed: 12/19/2022] Open
Abstract
Cells used in adoptive cell-transfer immunotherapies against cancer include dendritic cells (DCs), natural-killer cells, and CD8+ T-cells. These cells may have limited efficacy due to their lifespan, activity, and immunosuppressive effects of tumor cells. Therefore, increasing longevity and activity of these cells may boost their efficacy. Four cytokines that can extend immune effector-cell longevity are IL-2, IL-7, IL-21, and IL-15. This review will discuss current knowledge on effector-cell lifespans and the mechanisms by which IL-2, IL-7, IL-15, and IL-21 can extend effector-cell longevity. We will also discuss how lifespan and efficacy of these cells can be regulated to allow optimal clinical benefits.
Collapse
Affiliation(s)
- Sandeep Nayar
- MRC Centre for Transplantation; Kings College London; Guys Hospital ; London, UK
| | - Prokar Dasgupta
- MRC Centre for Transplantation; Kings College London; Guys Hospital ; London, UK
| | - Christine Galustian
- MRC Centre for Transplantation; Kings College London; Guys Hospital ; London, UK
| |
Collapse
|
44
|
Pilot Trial of FANG Immunotherapy in Ewing's Sarcoma. Mol Ther 2015; 23:1103-1109. [PMID: 25917459 PMCID: PMC4817748 DOI: 10.1038/mt.2015.43] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/06/2015] [Indexed: 12/18/2022] Open
Abstract
We report on 12 consecutive patients with advanced/metastatic Ewing's sarcoma who were treated as a separate cohort of a phase 1 trial of FANG autologous immunotherapy (1 × 10(6)-2.5 × 10(7) cells/intradermal injection each month for minimum 4 months). Safety and clinical response were monitored. Patient immune response to unmodified autologous tumor cells was assessed by gamma interferon-enzyme-linked immunospot (γIFN-ELISPOT) assay using peripheral blood mononuclear cells from baseline (pretreatment) and multiple postvaccination time points. None of the 12 patients (47 vaccinations) developed grade 2/3/4 drug-related toxicity. Median product release granulocyte-macrophage colony-stimulating factor expression was 1,941 pg/10(6) cells, and TGFβ1and TGFβ2 knockdown were 99 and 100%, respectively. Eight patients were assessed for ELISPOT response to autologous tumor cells at baseline and all (100%) were negative. In contrast, follow-up ELISPOT response at month 1 or month 4 (one patient) after FANG was positive in all eight patients. One patient achieved a partial tumor response (38% tumor reduction, RECIST 1.1). The Kaplan-Meier estimated survival of these 12 patients at 1 year was 75%. In this phase 1 study in patients with Ewing's sarcoma, FANG immunotherapy was well tolerated, elicited a tumor-specific systemic immune response in all patients, and was associated with favorable 1-year survival. Further clinical testing is indicated.
Collapse
|
45
|
Constitutive but not inducible attenuation of transforming growth factor β signaling increases natural killer cell responses without directly affecting dendritic cells early after persistent viral infection. J Virol 2015; 89:3343-55. [PMID: 25589641 DOI: 10.1128/jvi.03076-14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED Rapid innate responses to viral encounters are crucial to shaping the outcome of infection, from viral clearance to persistence. Transforming growth factor β (TGF-β) is a potent immune suppressor that is upregulated early upon viral infection and maintained during chronic infections in both mice and humans. However, the role of TGF-β signaling in regulating individual cell types in vivo is still unclear. Using infections with two different persistent viruses, murine cytomegalovirus (MCMV) and lymphocytic choriomeningitis virus (LCMV; Cl13), in their natural rodent host, we observed that TGF-β signaling on dendritic cells (DCs) did not dampen DC maturation or cytokine production in the early stages of chronic infection with either virus in vivo. In contrast, TGF-β signaling prior to (but not during) chronic viral infection directly restricted the natural killer (NK) cell number and effector function. This restriction likely compromised both the early control of and host survival upon MCMV infection but not the long-term control of LCMV infection. These data highlight the context and timing of TGF-β signaling on different innate cells that contribute to the early host response, which ultimately influences the outcome of chronic viral infection in vivo. IMPORTANCE In vivo host responses to pathogens are complex processes involving the cooperation of many different immune cells migrating to specific tissues over time, but these events cannot be replicated in vitro. Viruses causing chronic infections are able to subvert this immune response and represent a human health burden. Here we used two well-characterized viruses that are able to persist in their natural mouse host to dissect the role of the suppressive molecule TGF-β in dampening host responses to infection in vivo. This report presents information that allows an increased understanding of long-studied TGF-β signaling by examining its direct effect on different immune cells that are activated very early after in vivo viral infection and may aid with the development of new antiviral therapeutic strategies.
Collapse
|
46
|
Papageorgis P, Stylianopoulos T. Role of TGFβ in regulation of the tumor microenvironment and drug delivery (review). Int J Oncol 2015; 46:933-43. [PMID: 25573346 PMCID: PMC4306018 DOI: 10.3892/ijo.2015.2816] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 10/30/2014] [Indexed: 02/07/2023] Open
Abstract
Deregulation of cell signaling homeostasis is a predominant feature of cancer initiation and progression. Transforming growth factor β (TGFβ) is a pleiotropic cytokine, which regulates numerous biological processes of various tissues in an autocrine and paracrine manner. Aberrant activity of TGFβ signaling is well known to play dual roles in cancer, depending on tumor stage and cellular context. The crucial roles of TGFβ in modulating the tumor microenvironment, its contribution to the accumulation of mechanical forces within the solid constituents of a tumor and its effects on the effective delivery of drugs are also becoming increasingly clear. In this review, we discuss the latest advances in the efforts to unravel the effects of TGFβ signaling in various components of the tumor microenvironment and how these influence the generation of forces and the efficacy of drugs. We also report the implications of tumor mechanics in cancer therapy and the potential usage of anti-TGFβ agents to enhance drug delivery and augment existing therapeutic approaches. These findings provide new insights towards the significance of targeting TGFβ pathway to enhance personalized tumor treatment.
Collapse
Affiliation(s)
- Panagiotis Papageorgis
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus
| |
Collapse
|
47
|
Sparber F, Tripp CH, Komenda K, Scheffler JM, Clausen BE, Huber LA, Romani N, Stoitzner P. The late endosomal adaptor molecule p14 (LAMTOR2) regulates TGFβ1-mediated homeostasis of Langerhans cells. J Invest Dermatol 2015; 135:119-129. [PMID: 25078666 PMCID: PMC4285575 DOI: 10.1038/jid.2014.324] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 06/18/2014] [Accepted: 06/30/2014] [Indexed: 11/09/2022]
Abstract
Langerhans cells (LCs), a sub-population of dendritic cells (DCs) in the skin, participate in the regulation of immunity and peripheral tolerance. The adaptor molecule p14 is part of the late endosomal/lysosomal adaptor and mitogen-activated protein kinase and mammalian target of rapamycin (mTOR) activator/regulator (LAMTOR) complex, which mediates the activation of lysosome-associated extracellular signaling-regulated kinase (ERK) and the mTOR cascade. In previous work, we demonstrated that CD11c-specific deficiency of p14 disrupts LC homeostasis by affecting the LAMTOR-mediated ERK and mTOR signaling. In this study, we extended our analysis on p14 deficiency specifically in LCs. Langerin-specific ablation of p14 caused a complete loss of LCs, accompanied by an increased maturational phenotype of LCs. The absence of LCs in p14-deficient mice reduced contact hypersensitivity (CHS) responses to the contact sensitizer trinitrochlorobenzene. Analysis using bone marrow-derived DCs (BMDCs) revealed that p14 deficiency in DCs/LCs interfered with the LC-relevant transforming growth factor β1 (TGFβ1) pathway, by lowering TGFβ receptor II expression on BMDCs and LCs, as well as surface binding of TGFβ1 on BMDCs. We conclude that p14 deficiency affects TGFβ1 sensitivity of LCs, which is mandatory for their homeostasis and subsequently for their immunological function during CHS.
Collapse
Affiliation(s)
- Florian Sparber
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria
| | - Christoph H Tripp
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria
| | - Kerstin Komenda
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria
| | | | - Björn E Clausen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Lukas A Huber
- Division of Cell Biology, Biocenter Innsbruck, Innsbruck, Austria
| | - Nikolaus Romani
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria
| | - Patrizia Stoitzner
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria.
| |
Collapse
|
48
|
Dewitte H, Verbeke R, Breckpot K, Vandenbroucke RE, Libert C, De Smedt SC, Lentacker I. Choose your models wisely: How different murine bone marrow-derived dendritic cell protocols influence the success of nanoparticulate vaccines in vitro. J Control Release 2014; 195:138-46. [DOI: 10.1016/j.jconrel.2014.06.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 06/04/2014] [Accepted: 06/15/2014] [Indexed: 11/30/2022]
|
49
|
Sedger LM, McDermott MF. TNF and TNF-receptors: From mediators of cell death and inflammation to therapeutic giants - past, present and future. Cytokine Growth Factor Rev 2014; 25:453-72. [PMID: 25169849 DOI: 10.1016/j.cytogfr.2014.07.016] [Citation(s) in RCA: 560] [Impact Index Per Article: 50.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tumor Necrosis Factor (TNF), initially known for its tumor cytotoxicity, is a potent mediator of inflammation, as well as many normal physiological functions in homeostasis and health, and anti-microbial immunity. It also appears to have a central role in neurobiology, although this area of TNF biology is only recently emerging. Here, we review the basic biology of TNF and its normal effector functions, and discuss the advantages and disadvantages of therapeutic neutralization of TNF - now a commonplace practice in the treatment of a wide range of human inflammatory diseases. With over ten years of experience, and an emerging range of anti-TNF biologics now available, we also review their modes of action, which appear to be far more complex than had originally been anticipated. Finally, we highlight the current challenges for therapeutic intervention of TNF: (i) to discover and produce orally delivered small molecule TNF-inhibitors, (ii) to specifically target selected TNF producing cells or individual (diseased) tissue targets, and (iii) to pre-identify anti-TNF treatment responders. Although the future looks bright, the therapeutic modulation of TNF now moves into the era of personalized medicine with society's challenging expectations of durable treatment success and of achieving long-term disease remission.
Collapse
Affiliation(s)
- Lisa M Sedger
- Australian School of Advanced Medicine, Macquarie University, North Ryde, NSW 2109, Australia; The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 0200, Australia.
| | - Michael F McDermott
- Experimental Rheumatology, National Institute for Health Research - Leeds Musculoskeletal Biomedical Research Unit (NIHR-LMBRU), and Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Wellcome Trust Brenner Building, St James University, Beckett Street, West Yorkshire, Leeds LS9 7TF, UK.
| |
Collapse
|
50
|
Sun X, Robertson SA, Ingman WV. Regulation of epithelial cell turnover and macrophage phenotype by epithelial cell-derived transforming growth factor beta1 in the mammary gland. Cytokine 2013; 61:377-88. [PMID: 23290315 DOI: 10.1016/j.cyto.2012.12.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 11/07/2012] [Accepted: 12/04/2012] [Indexed: 12/31/2022]
Abstract
Transforming growth factor beta1 (TGFB1) is a multi-functional cytokine that regulates cell proliferation, apoptosis and immune system responses. In the breast, the mammary epithelium is the primary source of TGFB1 and increased expression is associated with increased breast cancer risk. This study was conducted to investigate the roles of epithelial cell-derived TGFB1 in regulation of epithelial cell activity and macrophage phenotype in the mammary gland. Tgfb1 null mutant and wildtype mammary epithelium was transplanted into contra-lateral sides of the cleared mammary gland of TGFB1 replete scid mice. Transplanted tissue was analysed for markers of proliferation and apoptosis to determine the effect of Tgfb1 null mutation on epithelial cell turnover, and was analysed by immunohistochemistry to investigate the location, abundance and phenotype of macrophages. The number of proliferating and dying ductal epithelial cells, determined by BrdU and TUNEL, was increased by 35% and 3.3-fold respectively in mammary gland transplanted with Tgfb1 null epithelium compared to wildtype epithelium (p<0.05). Abundance of F4/80+ macrophages in between Tgfb1 null epithelial cells compared to wildtype epithelial cells was increased by 50%. The number of iNOS+ and CCR7+ cells in the stroma surrounding Tgfb1 null alveolar epithelium was increased by 78% and 2-fold respectively, and dendriform MHC class II+ cells within ductal epithelium were decreased by 30%. We conclude that epithelial cell-derived TGFB1 in the mammary gland has two functions: (1) regulation of cellular turnover of epithelial cells, and (2) regulation of local macrophage phenotype. These findings shed new light on the diversity of roles of TGFB1 in the mammary gland which are likely to impact on breast cancer risk.
Collapse
Affiliation(s)
- Xuan Sun
- School of Paediatrics and Reproductive Health, University of Adelaide, Australia.
| | | | | |
Collapse
|