151
|
Waldner MJ, Neurath MF. TGFβ and the Tumor Microenvironment in Colorectal Cancer. Cells 2023; 12:1139. [PMID: 37190048 PMCID: PMC10137236 DOI: 10.3390/cells12081139] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/29/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Growing evidence supports an important role of the tumor microenvironment (TME) in the pathogenesis of colorectal cancer (CRC). Resident cells such as fibroblasts or immune cells infiltrating into the TME maintain continuous crosstalk with cancer cells and thereby regulate CRC progression. One of the most important molecules involved is the immunoregulatory cytokine transforming growth factor-β (TGFβ). TGFβ is released by various cells in the TME, including macrophages and fibroblasts, and it modulates cancer cell growth, differentiation, and cell death. Mutations in components of the TGF pathway, including TGFβ receptor type 2 or SMAD4, are among the most frequently detected mutations in CRC and have been associated with the clinical course of disease. Within this review, we will discuss our current understanding about the role of TGFβ in the pathogenesis of CRC. This includes novel data on the molecular mechanisms of TGFβ signaling in TME, as well as possible strategies for CRC therapy targeting the TGFβ pathway, including potential combinations with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Maximilian J. Waldner
- Department of Internal Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Markus F. Neurath
- Department of Internal Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| |
Collapse
|
152
|
Zhao Y, Chen X, Lin Y, Li Z, Su X, Fan S, Chen Y, Wang X, Liang G. USP25 inhibits renal fibrosis by regulating TGFβ-SMAD signaling pathway in Ang II-induced hypertensive mice. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166713. [PMID: 37059312 DOI: 10.1016/j.bbadis.2023.166713] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/22/2023] [Accepted: 04/02/2023] [Indexed: 04/16/2023]
Abstract
Renal fibrosis is a crucial pathological feature of hypertensive renal disease (HRD). In-depth analysis of the pathogenesis of fibrosis is of great significance for the development of new drugs for the treatment of HRD. USP25 is a deubiquitinase that can regulate the progression of many diseases, but its function in the kidney remains unclear. We found that USP25 was significantly increased in human and mice HRD kidney tissues. In the HRD model induced by Ang II, USP25-/- mice showed significant aggravation of renal dysfunction and fibrosis compared with the control mice. Consistently, AAV9-mediated overexpression of USP25 significantly improved renal dysfunction and fibrosis. Mechanistically, USP25 inhibited the TGF-β pathway by reducing SMAD4 K63-linked polyubiquitination, thereby suppressing SMAD2 nuclear translocation. In conclusion, this study demonstrates for the first time that the deubiquitinase USP25 plays an important regulatory role in HRD.
Collapse
Affiliation(s)
- Ying Zhao
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Xi Chen
- Department of Pharmacology, Medical College, Taizhou University, Taizhou, Jiaojiang 318000, Zhejiang, China
| | - Yimin Lin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhongding Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xian Su
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Shijie Fan
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yanghao Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xu Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China.
| |
Collapse
|
153
|
Mousset A, Lecorgne E, Bourget I, Lopez P, Jenovai K, Cherfils-Vicini J, Dominici C, Rios G, Girard-Riboulleau C, Liu B, Spector DL, Ehmsen S, Renault S, Hego C, Mechta-Grigoriou F, Bidard FC, Terp MG, Egeblad M, Gaggioli C, Albrengues J. Neutrophil extracellular traps formed during chemotherapy confer treatment resistance via TGF-β activation. Cancer Cell 2023; 41:757-775.e10. [PMID: 37037615 PMCID: PMC10228050 DOI: 10.1016/j.ccell.2023.03.008] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 12/13/2022] [Accepted: 03/07/2023] [Indexed: 04/12/2023]
Abstract
Metastasis is the major cause of cancer death, and the development of therapy resistance is common. The tumor microenvironment can confer chemotherapy resistance (chemoresistance), but little is known about how specific host cells influence therapy outcome. We show that chemotherapy induces neutrophil recruitment and neutrophil extracellular trap (NET) formation, which reduces therapy response in mouse models of breast cancer lung metastasis. We reveal that chemotherapy-treated cancer cells secrete IL-1β, which in turn triggers NET formation. Two NET-associated proteins are required to induce chemoresistance: integrin-αvβ1, which traps latent TGF-β, and matrix metalloproteinase 9, which cleaves and activates the trapped latent TGF-β. TGF-β activation causes cancer cells to undergo epithelial-to-mesenchymal transition and correlates with chemoresistance. Our work demonstrates that NETs regulate the activities of neighboring cells by trapping and activating cytokines and suggests that chemoresistance in the metastatic setting can be reduced or prevented by targeting the IL-1β-NET-TGF-β axis.
Collapse
Affiliation(s)
- Alexandra Mousset
- University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Enora Lecorgne
- University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France; University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), 3D-Hub-S Facility, Nice, France
| | - Isabelle Bourget
- University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France; University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), 3D-Hub-S Facility, Nice, France
| | - Pascal Lopez
- University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Kitti Jenovai
- University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Julien Cherfils-Vicini
- University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Chloé Dominici
- University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Géraldine Rios
- University Côte d'Azur, CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Sophia Antipolis, France
| | - Cédric Girard-Riboulleau
- University Côte d'Azur, CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Sophia Antipolis, France
| | - Bodu Liu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - David L Spector
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Sidse Ehmsen
- Department of Oncology, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Shufang Renault
- Circulating Tumor Biomarkers Laboratory, INSERM CIC-BT 1428, Department of Translational Research, Institut Curie, Paris, France
| | - Caroline Hego
- Circulating Tumor Biomarkers Laboratory, INSERM CIC-BT 1428, Department of Translational Research, Institut Curie, Paris, France
| | - Fatima Mechta-Grigoriou
- Stress and Cancer Laboratory, Institut Curie, INSERM, U830, PSL Research University, Ligue Nationale Contre le Cancer labeled Team, 26, Rue d'Ulm, 75005, Paris, France
| | - François-Clément Bidard
- Circulating Tumor Biomarkers Laboratory, INSERM CIC-BT 1428, Department of Translational Research, Institut Curie, Paris, France; Department of Medical Oncology, Institut Curie, Saint Cloud, Paris, France; University of Versailles Saint-Quentin-en-Yvelines (UVSQ), Paris-Saclay University, Saint Cloud, France
| | - Mikkel Green Terp
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Mikala Egeblad
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Cédric Gaggioli
- University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France; University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), 3D-Hub-S Facility, Nice, France.
| | - Jean Albrengues
- University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France.
| |
Collapse
|
154
|
Mannino F, Imbesi C, Bitto A, Minutoli L, Squadrito F, D'Angelo T, Booz C, Pallio G, Irrera N. Anti-oxidant and anti-inflammatory effects of ellagic and punicic acid in an in vitro model of cardiac fibrosis. Biomed Pharmacother 2023; 162:114666. [PMID: 37030134 DOI: 10.1016/j.biopha.2023.114666] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/10/2023] Open
Abstract
Cardiac fibrosis is a pathological process characterized by an excessive deposition of extracellular matrix (ECM) and an increased production of fibrillar collagen in the cardiac interstitium, mainly caused by the activation of cardiac fibroblasts and their transition into myofibroblasts. Oxidative stress is deeply implicated in the pathogenesis of cardiac fibrosis both directly and via its involvement in the tumor growth factor β1 (TGF-β1) signaling. Ellagic acid (EA) and punicic acid (PA) are the main components of the Punica granatum L (pomegranate) fruit and seed oil respectively, whose antioxidant, anti-inflammatory and anti-fibrotic effects have been previously described. Therefore, the aim of this study was to investigate the effects of EA or PA or EA+PA in an in vitro model of cardiac fibrosis. Immortalized Human Cardiac Fibroblasts (IM-HCF) were stimulated with 10 ng/ml of TGF-β1 for 24 h to induce a fibrotic damage. Cells were then treated with EA (1 µM), PA (1 µM) or EA+PA for additional 24 h. Both EA and PA reduced the pro-fibrotic proteins expressions and the intracellular reactive oxygen species (ROS) accumulation. The anti-oxidant activity was also observed by Nrf2 activation with the consequent TGF-β1-Smad2/3-MMP2/9 and Wnt/β-catenin signaling inhibition, thus reducing collagen production. EA and PA significantly inhibit NF-κB pathway and, consequently, TNF-α, IL-1β and IL-6 levels: the greater effect was observed when EA and PA were used in combination. These results suggest that EA, PA and in particular EA+PA might be effective in reducing fibrosis through their antioxidant and anti-inflammatory properties by the modulation of different molecular pathways.
Collapse
Affiliation(s)
- Federica Mannino
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy
| | - Chiara Imbesi
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy
| | - Letteria Minutoli
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy
| | - Francesco Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy
| | - Tommaso D'Angelo
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via C. Valeria Gazzi, 98125 Messina, Italy
| | - Christian Booz
- Division of Experimental Imaging, Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Giovanni Pallio
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy.
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy
| |
Collapse
|
155
|
Choi S, Cho N, Kim KK. The implications of alternative pre-mRNA splicing in cell signal transduction. Exp Mol Med 2023; 55:755-766. [PMID: 37009804 PMCID: PMC10167241 DOI: 10.1038/s12276-023-00981-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 01/05/2023] [Accepted: 01/27/2023] [Indexed: 04/04/2023] Open
Abstract
Cells produce multiple mRNAs through alternative splicing, which ensures proteome diversity. Because most human genes undergo alternative splicing, key components of signal transduction pathways are no exception. Cells regulate various signal transduction pathways, including those associated with cell proliferation, development, differentiation, migration, and apoptosis. Since proteins produced through alternative splicing can exhibit diverse biological functions, splicing regulatory mechanisms affect all signal transduction pathways. Studies have demonstrated that proteins generated by the selective combination of exons encoding important domains can enhance or attenuate signal transduction and can stably and precisely regulate various signal transduction pathways. However, aberrant splicing regulation via genetic mutation or abnormal expression of splicing factors negatively affects signal transduction pathways and is associated with the onset and progression of various diseases, including cancer. In this review, we describe the effects of alternative splicing regulation on major signal transduction pathways and highlight the significance of alternative splicing.
Collapse
Affiliation(s)
- Sunkyung Choi
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Namjoon Cho
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Kee K Kim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
156
|
Kang BN, Kang HJ, Kim S, Lee J, Lee J, Jeong HJ, Jeon S, Shin Y, Yoon C, Han C, Seo J, Yun J. Synthesis and biological evaluation of N-(3-fluorobenzyl)-4-(1-(methyl-d 3)-1H-indazol-5-yl)-5-(6-methylpyridin-2-yl)-1H-imidazol-2-amine as a novel, potent ALK5 receptor inhibitor. Bioorg Med Chem Lett 2023; 85:129205. [PMID: 36858078 DOI: 10.1016/j.bmcl.2023.129205] [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: 01/25/2023] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023]
Abstract
Specific inhibition of ALK5 provides a novel method for controlling the development of cancers and fibrotic diseases. In this work, a novel series of N-(3-fluorobenzyl)-4-(1-(methyl-d3)-1H-indazol-5-yl)-5-(6-methylpyridin-2-yl)-1H-imidazol-2-amine (11), a potential clinical candidate, was synthesized by strategic incorporation of deuterium at potential metabolic soft spots and identified as ALK5 inhibitors. This compound has a low potential for CYP-mediated drug-drug interactions as a CYP450 inhibitor (IC50 = >10 μM) and showed potent inhibitory effects in cellular assay (IC50 = 3.5 ± 0.4 nM). The pharmacokinetic evaluation of 11 in mice demonstrated moderate clearance (29.0 mL/min/kg) and also revealed high oral bioavailability in mice (F = 67.6%).
Collapse
Affiliation(s)
- Byung-Nam Kang
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea; Department of Chemistry, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hong-Jun Kang
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea
| | - Sunjoo Kim
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea
| | - Jungwoo Lee
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea
| | - Jinwoo Lee
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea
| | - Hee-Jin Jeong
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea
| | - Seeun Jeon
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea
| | - Youngdo Shin
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea
| | - Cheolhwan Yoon
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea
| | - Cheolkyu Han
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea; Department of Medical and Bioscience, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Jeongbeob Seo
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea.
| | - Jaesook Yun
- Department of Chemistry, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
157
|
Barlesi F, Isambert N, Felip E, Cho BC, Lee DH, Peguero J, Jerusalem G, Penel N, Saada-Bouzid E, Garrido P, Helwig C, Locke G, Ojalvo LS, Gulley JL. Bintrafusp Alfa, a Bifunctional Fusion Protein Targeting TGF-β and PD-L1, in Patients With Non-Small Cell Lung Cancer Resistant or Refractory to Immune Checkpoint Inhibitors. Oncologist 2023; 28:258-267. [PMID: 36571770 PMCID: PMC10020814 DOI: 10.1093/oncolo/oyac253] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 11/01/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Bintrafusp alfa is a first-in-class bifunctional fusion protein composed of the extracellular domain of transforming growth factor beta receptor II (a TGF-β "trap") fused to a human immunoglobulin G1 monoclonal antibody blocking programmed cell death 1 ligand 1 (PD-L1). We report the efficacy and safety in patients with non-small cell lung cancer (NSCLC) that progressed following anti-PD-(L)1 therapy. MATERIALS AND METHODS In this expansion cohort of NCT02517398-a global, open-label, phase I trial-adults with advanced NSCLC that progressed following chemotherapy and was primary refractory or had acquired resistance to anti-PD-(L)1 treatment received intravenous bintrafusp alfa 1200 mg every 2 weeks until confirmed progression, unacceptable toxicity, or trial withdrawal. The primary endpoint was best overall response (by Response Evaluation Criteria in Solid Tumors version 1.1 adjudicated by independent review committee); secondary endpoints included safety. RESULTS Eighty-three eligible patients (62 [74.7%] treated with ≥3 prior therapies) received bintrafusp alfa. Four patients (3 primary refractory, 1 acquired resistant) had confirmed partial responses (objective response rate, 4.8%; 95% CI, 1.3%-11.9%), and 9 had stable disease. Tumor cell PD-L1 expression was not associated with response. Nineteen patients (22.9%) experienced grade ≥3 treatment-related adverse events, most commonly asthenia (3 [3.6%]) and fatigue, eczema, and pruritus (2 each [2.4%]). One patient had grade 4 amylase increased. One patient died during treatment for pneumonia before initiation of bintrafusp alfa. CONCLUSION Although the primary endpoint was not met, bintrafusp alfa showed some clinical activity and a manageable safety profile in patients with heavily pretreated NSCLC, including prior anti-PD-(L)1 therapy. Tumor responses occurred irrespective of whether disease was primary refractory or had acquired resistance to prior anti-PD-(L)1 therapy.
Collapse
Affiliation(s)
- Fabrice Barlesi
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Nicolas Isambert
- Service d’oncologie médicale CLCC Georges-François Leclerc, Dijon, France
| | - Enriqueta Felip
- Oncology Department, Vall d’Hebron University Hospital and Institute of Oncology (VHIO), UVic-UCC, IOB-Quiron, Barcelona, Spain
| | - Byoung Chul Cho
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dae Ho Lee
- Department of Oncology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Julio Peguero
- Department of Research, Oncology Consultants, Houston, TX, USA
| | - Guy Jerusalem
- Medical Oncology, CHU Sart Tilman Liege and Liege University, Domaine Universitaire, Liege, Belgium
| | - Nicolas Penel
- Department of Medical Oncology, Lille University, Medical School and Centre Oscar Lambret, Lille, France
| | - Esma Saada-Bouzid
- Department of Medical Oncology, Early Phase Trials Unit, Centre Antoine Lacassagne, Nice, France
| | - Pilar Garrido
- Lung Cancer Unit, University Hospital Ramón y Cajal (IRYCIS), Medical Oncology Department, Madrid, Spain
| | | | | | | | - James L Gulley
- Corresponding author: James L. Gulley, MD, PhD, Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, 10 Center Drive, 13N240, Bethesda, MD 20892, USA. Tel: +1 301 480 7164; Fax: +1 301 480 6288;
| |
Collapse
|
158
|
Yang L, Wan N, Gong F, Wang X, Feng L, Liu G. Transcription factors and potential therapeutic targets for pulmonary hypertension. Front Cell Dev Biol 2023; 11:1132060. [PMID: 37009479 PMCID: PMC10064017 DOI: 10.3389/fcell.2023.1132060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 03/03/2023] [Indexed: 03/19/2023] Open
Abstract
Pulmonary hypertension (PH) is a refractory and fatal disease characterized by excessive pulmonary arterial cell remodeling. Uncontrolled proliferation and hypertrophy of pulmonary arterial smooth muscle cells (PASMCs), dysfunction of pulmonary arterial endothelial cells (PAECs), and abnormal perivascular infiltration of immune cells result in pulmonary arterial remodeling, followed by increased pulmonary vascular resistance and pulmonary pressure. Although various drugs targeting nitric oxide, endothelin-1 and prostacyclin pathways have been used in clinical settings, the mortality of pulmonary hypertension remains high. Multiple molecular abnormalities have been implicated in pulmonary hypertension, changes in numerous transcription factors have been identified as key regulators in pulmonary hypertension, and a role for pulmonary vascular remodeling has been highlighted. This review consolidates evidence linking transcription factors and their molecular mechanisms, from pulmonary vascular intima PAECs, vascular media PASMCs, and pulmonary arterial adventitia fibroblasts to pulmonary inflammatory cells. These findings will improve the understanding of particularly interactions between transcription factor-mediated cellular signaling pathways and identify novel therapies for pulmonary hypertension.
Collapse
Affiliation(s)
- Liu Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Naifu Wan
- Department of Vascular & Cardiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fanpeng Gong
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xianfeng Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Lei Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Guizhu Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- *Correspondence: Guizhu Liu,
| |
Collapse
|
159
|
Ben Hamouda S, Essafi-Benkhadir K. Interplay between Signaling Pathways and Tumor Microenvironment Components: A Paradoxical Role in Colorectal Cancer. Int J Mol Sci 2023; 24:ijms24065600. [PMID: 36982677 PMCID: PMC10057671 DOI: 10.3390/ijms24065600] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023] Open
Abstract
The study of the tumor microenvironment (TME) has become an important part of colorectal cancer (CRC) research. Indeed, it is now accepted that the invasive character of a primary CRC is determined not only by the genotype of the tumor cells, but also by their interactions with the extracellular environment, which thereby orchestrates the development of the tumor. In fact, the TME cells are a double-edged sword as they play both pro- and anti-tumor roles. The interaction of the tumor-infiltrating cells (TIC) with the cancer cells induces the polarization of the TIC, exhibiting an antagonist phenotype. This polarization is controlled by a plethora of interconnected pro- and anti-oncogenic signaling pathways. The complexity of this interaction and the dual function of these different actors contribute to the failure of CRC control. Thus, a better understanding of such mechanisms is of great interest and provides new opportunities for the development of personalized and efficient therapies for CRC. In this review, we summarize the signaling pathways linked to CRC and their implication in the development or inhibition of the tumor initiation and progression. In the second part, we enlist the major components of the TME and discuss the complexity of their cells functions.
Collapse
|
160
|
Shi ZD, Pang K, Wu ZX, Dong Y, Hao L, Qin JX, Wang W, Chen ZS, Han CH. Tumor cell plasticity in targeted therapy-induced resistance: mechanisms and new strategies. Signal Transduct Target Ther 2023; 8:113. [PMID: 36906600 PMCID: PMC10008648 DOI: 10.1038/s41392-023-01383-x] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 12/07/2022] [Accepted: 02/20/2023] [Indexed: 03/13/2023] Open
Abstract
Despite the success of targeted therapies in cancer treatment, therapy-induced resistance remains a major obstacle to a complete cure. Tumor cells evade treatments and relapse via phenotypic switching driven by intrinsic or induced cell plasticity. Several reversible mechanisms have been proposed to circumvent tumor cell plasticity, including epigenetic modifications, regulation of transcription factors, activation or suppression of key signaling pathways, as well as modification of the tumor environment. Epithelial-to-mesenchymal transition, tumor cell and cancer stem cell formation also serve as roads towards tumor cell plasticity. Corresponding treatment strategies have recently been developed that either target plasticity-related mechanisms or employ combination treatments. In this review, we delineate the formation of tumor cell plasticity and its manipulation of tumor evasion from targeted therapy. We discuss the non-genetic mechanisms of targeted drug-induced tumor cell plasticity in various types of tumors and provide insights into the contribution of tumor cell plasticity to acquired drug resistance. New therapeutic strategies such as inhibition or reversal of tumor cell plasticity are also presented. We also discuss the multitude of clinical trials that are ongoing worldwide with the intention of improving clinical outcomes. These advances provide a direction for developing novel therapeutic strategies and combination therapy regimens that target tumor cell plasticity.
Collapse
Affiliation(s)
- Zhen-Duo Shi
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China.,Department of Urology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China.,School of Life Sciences, Jiangsu Normal University, Jiangsu, China.,Department of Urology, Heilongjiang Provincial Hospital, Heilongjiang, China
| | - Kun Pang
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China.,Department of Urology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Yang Dong
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China.,Department of Urology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Lin Hao
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China.,Department of Urology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Jia-Xin Qin
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China.,Department of Urology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Wei Wang
- Department of Medical College, Southeast University, Nanjing, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| | - Cong-Hui Han
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China. .,Department of Urology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China. .,School of Life Sciences, Jiangsu Normal University, Jiangsu, China. .,Department of Urology, Heilongjiang Provincial Hospital, Heilongjiang, China.
| |
Collapse
|
161
|
Cong Y, Wang Y, Yuan T, Zhang Z, Ge J, Meng Q, Li Z, Sun S. Macrophages in aseptic loosening: Characteristics, functions, and mechanisms. Front Immunol 2023; 14:1122057. [PMID: 36969165 PMCID: PMC10030580 DOI: 10.3389/fimmu.2023.1122057] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/13/2023] [Indexed: 03/10/2023] Open
Abstract
Aseptic loosening (AL) is the most common complication of total joint arthroplasty (TJA). Both local inflammatory response and subsequent osteolysis around the prosthesis are the fundamental causes of disease pathology. As the earliest change of cell behavior, polarizations of macrophages play an essential role in the pathogenesis of AL, including regulating inflammatory responses and related pathological bone remodeling. The direction of macrophage polarization is closely dependent on the microenvironment of the periprosthetic tissue. When the classically activated macrophages (M1) are characterized by the augmented ability to produce proinflammatory cytokines, the primary functions of alternatively activated macrophages (M2) are related to inflammatory relief and tissue repair. Yet, both M1 macrophages and M2 macrophages are involved in the occurrence and development of AL, and a comprehensive understanding of polarized behaviors and inducing factors would help in identifying specific therapies. In recent years, studies have witnessed novel discoveries regarding the role of macrophages in AL pathology, the shifts between polarized phenotype during disease progression, as well as local mediators and signaling pathways responsible for regulations in macrophages and subsequent osteoclasts (OCs). In this review, we summarize recent progress on macrophage polarization and related mechanisms during the development of AL and discuss new findings and concepts in the context of existing work.
Collapse
Affiliation(s)
- Yehao Cong
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yi Wang
- Department of Joint Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Tao Yuan
- Department of Joint Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Zheng Zhang
- Department of Joint Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Jianxun Ge
- Department of Joint Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Qi Meng
- Department of Joint Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Ziqing Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- *Correspondence: Ziqing Li, ; Shui Sun,
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Joint Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- *Correspondence: Ziqing Li, ; Shui Sun,
| |
Collapse
|
162
|
Joo SH, Kim J, Hong J, Fakhraei Lahiji S, Kim YH. Dissolvable Self-Locking Microneedle Patches Integrated with Immunomodulators for Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2209966. [PMID: 36528846 DOI: 10.1002/adma.202209966] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/08/2022] [Indexed: 06/17/2023]
Abstract
Advancements in micro-resolution 3D printers have significantly facilitated the development of highly complex mass-producible drug delivery platforms. Conventionally, due to the limitations of micro-milling machineries, dissolvable microneedles (MNs) are mainly fabricated in cone-shaped geometry with limited drug delivery accuracy. Herein, to overcome the limitations of conventional MNs, a novel projection micro-stereolithography 3D printer-based self-locking MN for precise skin insertion, adhesion, and transcutaneous microdose drug delivery is presented. The geometry of self-locking MN consists of a sharp skin-penetrating tip, a wide skin interlocking body, and a narrow base with mechanical supports fabricated over a flexible hydrocolloid patch to improve the accuracy of skin penetration into irregular surfaces. Melanoma, a type of skin cancer, is selected as the model for the investigation of self-locking MNs due to its irregular and uneven surface. In vivo immunotherapy efficacy is evaluated by integrating SD-208, a novel transforming growth factor-β (TGF-β) inhibitor that suppresses the proliferation and metastasis of tumors, and anti-PD-L1 (aPD-L1 Ab), an immune checkpoint inhibitor that induces T cell-mediated tumor cell death, into self-locking MNs and comparing them with intratumoral injection. Evaluation of (aPD-L1 Ab)/SD-208 delivery effectiveness in B16F10 melanoma-bearing mice model confirms significantly improved dose efficacy of self-locking MNs compared with intratumoral injection.
Collapse
Affiliation(s)
- Seung-Hwan Joo
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seongdong-gu, Seoul, 04763, Republic of Korea
- Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Jaehyun Kim
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seongdong-gu, Seoul, 04763, Republic of Korea
- Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Juhyeong Hong
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seongdong-gu, Seoul, 04763, Republic of Korea
- Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Shayan Fakhraei Lahiji
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seongdong-gu, Seoul, 04763, Republic of Korea
- Cursus Bio Inc., Icure Tower, Gangnam-gu, Seoul, 06170, Republic of Korea
| | - Yong-Hee Kim
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seongdong-gu, Seoul, 04763, Republic of Korea
- Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seongdong-gu, Seoul, 04763, Republic of Korea
- Cursus Bio Inc., Icure Tower, Gangnam-gu, Seoul, 06170, Republic of Korea
| |
Collapse
|
163
|
Ali S, Rehman MU, Yatoo AM, Arafah A, Khan A, Rashid S, Majid S, Ali A, Ali MN. TGF-β signaling pathway: Therapeutic targeting and potential for anti-cancer immunity. Eur J Pharmacol 2023; 947:175678. [PMID: 36990262 DOI: 10.1016/j.ejphar.2023.175678] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 03/07/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023]
Abstract
Transforming growth factor-β (TGFβ) is a pleiotropic secretory cytokine exhibiting both cancer-inhibitory and promoting properties. It transmits its signals via Suppressor of Mother against Decapentaplegic (SMAD) and non-SMAD pathways and regulates cell proliferation, differentiation, invasion, migration, and apoptosis. In non-cancer and early-stage cancer cells, TGFβ signaling suppresses cancer progression via inducing apoptosis, cell cycle arrest, or anti-proliferation, and promoting cell differentiation. On the other hand, TGFβ may also act as an oncogene in advanced stages of tumors, wherein it develops immune-suppressive tumor microenvironments and induces the proliferation of cancer cells, invasion, angiogenesis, tumorigenesis, and metastasis. Higher TGFβ expression leads to the instigation and development of cancer. Therefore, suppressing TGFβ signals may present a potential treatment option for inhibiting tumorigenesis and metastasis. Different inhibitory molecules, including ligand traps, anti-sense oligo-nucleotides, small molecule receptor-kinase inhibitors, small molecule inhibitors, and vaccines, have been developed and clinically trialed for blocking the TGFβ signaling pathway. These molecules are not pro-oncogenic response-specific but block all signaling effects induced by TGFβ. Nonetheless, targeting the activation of TGFβ signaling with maximized specificity and minimized toxicity can enhance the efficacy of therapeutic approaches against this signaling pathway. The molecules that are used to target TGFβ are non-cytotoxic to cancer cells but designed to curtail the over-activation of invasion and metastasis driving TGFβ signaling in stromal and cancer cells. Here, we discussed the critical role of TGFβ in tumorigenesis, and metastasis, as well as the outcome and the promising achievement of TGFβ inhibitory molecules in the treatment of cancer.
Collapse
|
164
|
Rahavi H, Alizadeh-Navaei R, Tehrani M. Efficacy of therapies targeting TGF-β in solid tumors: a systematic review and meta-analysis of clinical trials. Immunotherapy 2023; 15:283-292. [PMID: 36789642 DOI: 10.2217/imt-2022-0079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
Aims: A comprehensive meta-analysis was conducted to explore the efficacy of TGF-β blockade therapies in solid tumors. Patients & methods: Results of overall survival (OS), progression-free survival (PFS), time to progression (TTP) and overall response rate (ORR) with their 95% CI were calculated. Also, subgroup analyses were conducted according to the categories of TGF-β blocker alone or combined with chemotherapy or radiotherapy. Results: Overall OS, PFS, TTP and ORR were 10.5 months (95% CI: 7.76-13.25), 2.54 months (95% CI: 1.66-3.43), 4.69 months (95% CI: 3.18-6.21) and 0.83% (95% CI: 0.82-0.85), respectively. Conclusion: Collectively, TGF-β blockade combined with chemotherapy or radiotherapy showed more favorable clinical outcomes than monotherapy using TGF-β blockade.
Collapse
Affiliation(s)
- Hossein Rahavi
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, 48471-91971, Iran
| | - Reza Alizadeh-Navaei
- Gastrointestinal Cancer Research Center, Non-communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, 48471-91971, Iran
| | - Mohsen Tehrani
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, 48471-91971, Iran.,Molecular & Cell Biology Research Center (MCBRC), Mazandaran University of Medical Sciences, Sari, 48471-91971, Iran
| |
Collapse
|
165
|
Huang Q, Liu M, Zhang D, Lin BB, Fu X, Zhang Z, Zhang B, Dong JT. Nitazoxanide inhibits acetylated KLF5-induced bone metastasis by modulating KLF5 function in prostate cancer. BMC Med 2023; 21:68. [PMID: 36810084 PMCID: PMC9945734 DOI: 10.1186/s12916-023-02763-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/30/2023] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND Castration-resistant prostate cancer often metastasizes to the bone, and such bone metastases eventually become resistant to available therapies, leading to the death of patients. Enriched in the bone, TGF-β plays a pivotal role in bone metastasis development. However, directly targeting TGF-β or its receptors has been challenging for the treatment of bone metastasis. We previously found that TGF-β induces and then depends on the acetylation of transcription factor KLF5 at K369 to regulate multiple biological processes, including the induction of EMT, cellular invasiveness, and bone metastasis. Acetylated KLF5 (Ac-KLF5) and its downstream effectors are thus potential therapeutic targets for treating TGF-β-induced bone metastasis in prostate cancer. METHODS A spheroid invasion assay was applied to prostate cancer cells expressing KLF5K369Q, which mimics Ac-KLF5, to screen 1987 FDA-approved drugs for invasion suppression. Luciferase- and KLF5K369Q-expressing cells were injected into nude mice via the tail artery to model bone metastasis. Bioluminescence imaging, micro-CT), and histological analyses were applied to monitor and evaluate bone metastases. RNA-sequencing, bioinformatic, and biochemical analyses were used to understand nitazoxanide (NTZ)-regulated genes, signaling pathways, and the underlying mechanisms. The binding of NTZ to KLF5 proteins was evaluated using fluorescence titration, high-performance liquid chromatography (HPLC), and circular dichroism (CD) analysis. RESULTS NTZ, an anthelmintic agent, was identified as a potent invasion inhibitor in the screening and validation assays. In KLF5K369Q-induced bone metastasis, NTZ exerted a potent inhibitory effect in preventive and therapeutic modes. NTZ also inhibited osteoclast differentiation, a cellular process responsible for bone metastasis induced by KLF5K369Q. NTZ attenuated the function of KLF5K369Q in 127 genes' upregulation and 114 genes' downregulation. Some genes' expression changes were significantly associated with worse overall survival in patients with prostate cancer. One such change was the upregulation of MYBL2, which functionally promotes bone metastasis in prostate cancer. Additional analyses demonstrated that NTZ bound to the KLF5 protein, KLF5K369Q bound to the promoter of MYBL2 to activate its transcription, and NTZ attenuated the binding of KLF5K369Q to the MYBL2 promoter. CONCLUSIONS NTZ is a potential therapeutic agent for bone metastasis induced by the TGF-β/Ac-KLF5 signaling axis in prostate cancer and likely other cancers.
Collapse
Affiliation(s)
- Qingqing Huang
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Blvd, Shenzhen, 518055, China
| | - Mingcheng Liu
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Blvd, Shenzhen, 518055, China
| | - Duo Zhang
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Blvd, Shenzhen, 518055, China
| | - Bing-Biao Lin
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Blvd, Shenzhen, 518055, China.,Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, China
| | - Xing Fu
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Blvd, Shenzhen, 518055, China
| | - Zhiqian Zhang
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Blvd, Shenzhen, 518055, China
| | - Baotong Zhang
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Blvd, Shenzhen, 518055, China
| | - Jin-Tang Dong
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Blvd, Shenzhen, 518055, China.
| |
Collapse
|
166
|
Shao CG, Sinha NR, Mohan RR, Webel AD. Novel Therapies for the Prevention of Fibrosis in Glaucoma Filtration Surgery. Biomedicines 2023; 11:657. [PMID: 36979636 PMCID: PMC10045591 DOI: 10.3390/biomedicines11030657] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/07/2023] [Accepted: 02/15/2023] [Indexed: 02/24/2023] Open
Abstract
Conjunctival fibrosis remains the major impediment to the success of glaucoma filtration surgery. Anti-metabolites remain the gold standard for mitigating post-surgical fibrosis, but they are associated with high complication rates and surgical failure rates. Establishing a more targeted approach to attenuate conjunctival fibrosis may revolutionize the surgical approach to glaucoma. A new strategy is needed to prevent progressive tissue remodeling and formation of a fibrotic scar, subsequently increasing surgical success and reducing the prevalence of glaucoma-related vision loss. Advancements in our understanding of molecular signaling and biomechanical cues in the conjunctival tissue architecture are broadening the horizon for new therapies and biomaterials for the mitigation of fibrosis. This review aims to highlight the strategies and current state of promising future approaches for targeting fibrosis in glaucoma filtration surgery.
Collapse
Affiliation(s)
| | - Nishant R. Sinha
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65212, USA
- One-Health Vision Research Program, Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Rajiv R. Mohan
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65212, USA
- One-Health Vision Research Program, Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, USA
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Aaron D. Webel
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
167
|
Fish BL, Hart B, Gasperetti T, Narayanan J, Gao F, Veley D, Pierce L, Himburg HA, MacVittie T, Medhora M. IPW-5371 mitigates the delayed effects of acute radiation exposure in WAG/RijCmcr rats when started 15 days after PBI with bone marrow sparing. Int J Radiat Biol 2023; 99:1119-1129. [PMID: 36794325 PMCID: PMC10330589 DOI: 10.1080/09553002.2023.2173825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 02/17/2023]
Abstract
PURPOSE To test IPW-5371 for the mitigation of the delayed effects of acute radiation exposure (DEARE). Survivors of acute radiation exposure are at risk for developing delayed multi-organ toxicities; however, there are no FDA-approved medical countermeasures (MCM) to mitigate DEARE. METHODS WAG/RijCmcr female rat model of partial-body irradiation (PBI), by shielding part of one hind leg, was used to test IPW-5371 (7 and 20 mg kg-1 d-1) for mitigation of lung and kidney DEARE when started 15 d after PBI. Rats were fed known amounts of IPW-5371 using a syringe, instead of delivery by daily oral gavage, sparing exacerbation of esophageal injury by radiation. The primary endpoint, all-cause morbidity was assessed over 215 d. Secondary endpoints: body weight, breathing rate and blood urea nitrogen were also assessed. RESULTS IPW-5371 enhanced survival (primary endpoint) as well as attenuated secondary endpoints of lung and kidney injuries by radiation. CONCLUSION To provide a window for dosimetry and triage, as well as avoid oral delivery during the acute radiation syndrome (ARS), the drug regimen was started at 15 d after 13.5 Gy PBI. The experimental design to test mitigation of DEARE was customized for translation in humans, using an animal model of radiation that was designed to simulate a radiologic attack or accident. The results support advanced development of IPW-5371 to mitigate lethal lung and kidney injuries after irradiation of multiple organs.
Collapse
Affiliation(s)
- Brian L. Fish
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226
| | - Barry Hart
- Innovation Pathways, Palo Alto, CA, 94301
| | - Tracy Gasperetti
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226
| | - Jayashree Narayanan
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226
| | - Feng Gao
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226
| | - Dana Veley
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226
| | - Lauren Pierce
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226
| | - Heather A. Himburg
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226
| | - Thomas MacVittie
- Innovation Pathways, Palo Alto, CA, 94301
- Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD 21201
| | - Meetha Medhora
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226
| |
Collapse
|
168
|
Planta D, Gerwinn T, Salemi S, Horst M. Neurogenic Lower Urinary Tract Dysfunction in Spinal Dysraphism: Morphological and Molecular Evidence in Children. Int J Mol Sci 2023; 24:ijms24043692. [PMID: 36835106 PMCID: PMC9959703 DOI: 10.3390/ijms24043692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Spinal dysraphism, most commonly myelomeningocele, is the typical cause of a neurogenic lower urinary tract dysfunction (NLUTD) in childhood. The structural changes in the bladder wall in spinal dysraphism already occur in the fetal period and affect all bladder wall compartments. The progressive decrease in smooth muscle and the gradual increase in fibrosis in the detrusor, the impairment of the barrier function of the urothelium, and the global decrease in nerve density, lead to severe functional impairment characterized by reduced compliance and increased elastic modulus. Children present a particular challenge, as their diseases and capabilities evolve with age. An increased understanding of the signaling pathways involved in lower urinary tract development and function could also fill an important knowledge gap at the interface between basic science and clinical implications, leading to new opportunities for prenatal screening, diagnosis, and therapy. In this review, we aim to summarize the evidence on structural, functional, and molecular changes in the NLUTD bladder in children with spinal dysraphism and discuss possible strategies for improved management and for the development of new therapeutic approaches for affected children.
Collapse
Affiliation(s)
- Dafni Planta
- Division of Pediatric Urology, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
| | - Tim Gerwinn
- Division of Pediatric Urology, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
| | - Souzan Salemi
- Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Maya Horst
- Division of Pediatric Urology, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Correspondence:
| |
Collapse
|
169
|
The mechanism and therapy of aortic aneurysms. Signal Transduct Target Ther 2023; 8:55. [PMID: 36737432 PMCID: PMC9898314 DOI: 10.1038/s41392-023-01325-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 12/15/2022] [Accepted: 01/14/2023] [Indexed: 02/05/2023] Open
Abstract
Aortic aneurysm is a chronic aortic disease affected by many factors. Although it is generally asymptomatic, it poses a significant threat to human life due to a high risk of rupture. Because of its strong concealment, it is difficult to diagnose the disease in the early stage. At present, there are no effective drugs for the treatment of aneurysms. Surgical intervention and endovascular treatment are the only therapies. Although current studies have discovered that inflammatory responses as well as the production and activation of various proteases promote aortic aneurysm, the specific mechanisms remain unclear. Researchers are further exploring the pathogenesis of aneurysms to find new targets for diagnosis and treatment. To better understand aortic aneurysm, this review elaborates on the discovery history of aortic aneurysm, main classification and clinical manifestations, related molecular mechanisms, clinical cohort studies and animal models, with the ultimate goal of providing insights into the treatment of this devastating disease. The underlying problem with aneurysm disease is weakening of the aortic wall, leading to progressive dilation. If not treated in time, the aortic aneurysm eventually ruptures. An aortic aneurysm is a local enlargement of an artery caused by a weakening of the aortic wall. The disease is usually asymptomatic but leads to high mortality due to the risk of artery rupture.
Collapse
|
170
|
Chen M, Guo X, Zhong Y, Liu Y, Cai B, Wu R, Huang C, Zhou C. AMH inhibits androgen production in human theca cells. J Steroid Biochem Mol Biol 2023; 226:106216. [PMID: 36356855 DOI: 10.1016/j.jsbmb.2022.106216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/09/2022]
Abstract
Both excessive ovarian production of AMH and androgen are important features of polycystic ovary syndrome (PCOS). Present study aimed to explore the direct effect of AMH on androgen production in human theca cells. Primary cultured human theca cells were treated with AMH, an ALK2 (the BMP type 1 receptor) inhibitor and an ALK5 (the TGFβ type 1 receptor) inhibitor. AMH significantly suppresses the expression of the androgen synthesis-related enzyme CYP17A1 and reduces the production of androstenedione and testosterone in normal human theca cells and PCOS theca cells. Inhibitors of ALK2/3 and ALK5 antagonize the effect of AMH on the expression of CYP17A1. Although both ALK5 and ALK2 interact with AMHR2 in the presence of AMH, AMH activated neither TGFβR-Smads (Smad 2/3) nor BMPR-Smads (Smad 1/5/8). Our data suggested that AMH suppresses androgen synthesis-related enzyme CYP17A1 expression and inhibits androgen production in human theca cells, which process may be mediated by ALK2 and ALK5.
Collapse
Affiliation(s)
- Minghui Chen
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Reproductive Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Xi Guo
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Reproductive Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yiping Zhong
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Reproductive Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yang Liu
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Reproductive Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bing Cai
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Reproductive Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Rihan Wu
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Reproductive Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chuan Huang
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Reproductive Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Canquan Zhou
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Reproductive Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
171
|
Cuevas RA, Wong R, Joolharzadeh P, Moorhead WJ, Chu CC, Callahan J, Crane A, Boufford CK, Parise AM, Parwal A, Behzadi P, St Hilaire C. Ecto-5'-nucleotidase (Nt5e/CD73)-mediated adenosine signaling attenuates TGFβ-2 induced elastin and cellular contraction. Am J Physiol Cell Physiol 2023; 324:C327-C338. [PMID: 36503240 PMCID: PMC9902218 DOI: 10.1152/ajpcell.00054.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 11/21/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
Arterial calcification due to deficiency of CD73 (ACDC) is a rare genetic disease caused by a loss-of-function mutation in the NT5E gene encoding the ecto-5'-nucleotidase (cluster of differentiation 73, CD73) enzyme. Patients with ACDC develop vessel arteriomegaly, tortuosity, and vascular calcification in their lower extremity arteries. Histological analysis shows that patients with ACDC vessels exhibit fragmented elastin fibers similar to that seen in aneurysmal-like pathologies. It is known that alterations in transforming growth factor β (TGFβ) pathway signaling contribute to this elastin phenotype in several connective tissue diseases, as TGFβ regulates extracellular matrix (ECM) remodeling. Our study investigates whether CD73-derived adenosine modifies TGFβ signaling in vascular smooth muscle cells (SMCs). We show that Nt5e-/- SMCs have elevated contractile markers and elastin gene expression compared with Nt5e+/+ SMCs. Ecto-5'-nucleotidase (Nt5e)-deficient SMCs exhibit increased TGFβ-2 and activation of small mothers against decapentaplegic (SMAD) signaling, elevated elastin transcript and protein, and potentiate SMC contraction. These effects were diminished when the A2b adenosine receptor was activated. Our results identify a novel link between adenosine and TGFβ signaling, where adenosine signaling via the A2b adenosine receptor attenuates TGFβ signaling to regulate SMC homeostasis. We discuss how disruption in adenosine signaling is implicated in ACDC vessel tortuosity and could potentially contribute to other aneurysmal pathogenesis.
Collapse
Affiliation(s)
- Rolando A Cuevas
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ryan Wong
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Pouya Joolharzadeh
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - William J Moorhead
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Claire C Chu
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jack Callahan
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alex Crane
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Camille K Boufford
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Angelina M Parise
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Aneesha Parwal
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Parya Behzadi
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Cynthia St Hilaire
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
172
|
Lai X, Huang S, Lin Y, Qiu Y, Pu L, Lin S, Zeng Q, Huang W, Wang Z. DACT2 protects against pulmonary fibrosis via suppressing glycolysis in lung myofibroblasts. Int J Biol Macromol 2023; 226:291-300. [PMID: 36481337 DOI: 10.1016/j.ijbiomac.2022.11.324] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/23/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive fibrotic lung disease with poor prognosis and few treatment options. Dapper homolog 2 (DACT2), a member of the DACT gene family, plays crucial roles in tissue development and injury. However, its functions and molecular mechanisms in IPF remain largely unknown. We aimed to investigate the role of DACT2 in the development of pulmonary fibrosis and the therapeutic potential of targeting DACT2 related signaling pathways. METHODS In our study, adeno-associated virus serotype 6 (AAV6)-mediated DACT2 overexpression was assessed in several mice models of experimental pulmonary fibrosis in vivo. The role of DACT2 in lung myofibroblast differentiation was determined by DACT2 overexpression in vitro. The glucose uptake, extracellular acidification rate, intracellular adenosine-triphosphate (ATP) level and lactate levels of myofibroblasts were detected after DACT2 overexpression. The LDHA degradation rate and colocalization with lysosomes were monitored as well. RESULTS Intratracheal administration of AAV6-mediated DACT2 overexpression apparently attenuated pulmonary fibrosis in experimental pulmonary fibrosis models. In vitro experiments revealed that DACT2 inhibited TGF-β-induced myofibroblast differentiation by promoting lysosome-mediated LDHA degradation and thus suppressing glycolysis in myofibroblasts. CONCLUSION In conclusion, our findings support for DACT2 as a novel pharmacological target for pulmonary fibrosis treatments.
Collapse
Affiliation(s)
- Xiaofan Lai
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shaojie Huang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yingying Lin
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuan Qiu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
| | - Lvya Pu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Sijia Lin
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qihao Zeng
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wenqi Huang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Zhongxing Wang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
173
|
Clark AG, Bertrand FE, Sigounas G. A potential requirement for Smad3 phosphorylation in Notch-mediated EMT in colon cancer. Adv Biol Regul 2023; 88:100957. [PMID: 36739740 DOI: 10.1016/j.jbior.2023.100957] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/04/2023] [Accepted: 01/27/2023] [Indexed: 01/30/2023]
Abstract
Colorectal cancer (CRC) remains a challenging disease to treat due to several factors including stemness and epithelial to mesenchymal transition (EMT). Dysfunctional signaling pathways such as Notch and TGF-β contribute to these phenomena. We previously found that cells expressing constitutively active Notch1 also had increased expression of Smad3, an important member of the TGF-β signaling pathway. We hypothesized that Smad3, mediates the Notch-induced stemness and EMT observed in CRC cells. The human colorectal carcinoma cell line HCT-116, stably transduced with constitutively active Notch-1 (ICN) or a GFP-vector control was treated with different combinations of TGF-β1, DAPT (a Notch inhibitor), or SIS3 (a Smad3 inhibitor). Western blot analysis was performed to determine the effects of Smad3 stimulation and inhibition on Notch and potential downstream EMT-related targets, CD44, Slug and Snail. Smad3 inhibition induced a decrease in Notch1 and Notch3 receptor expression and effectively inhibited CD44, Slug, and Snail expression. Colosphere forming ability was also reduced in cells with inhibited Smad3. These results indicate a key role of TGF-β signaling in Notch1-induced tumorigenesis, and suggest a potential use for Smad3 inhibitors in combination with Notch1 inhibitors that are already in use for CRC treatments.
Collapse
Affiliation(s)
- Alexander G Clark
- Department of Internal Medicine, Division of Hematology/Oncology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Fred E Bertrand
- Department of Clinical and Diagnostic Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - George Sigounas
- Department of Internal Medicine, Division of Hematology/Oncology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA.
| |
Collapse
|
174
|
Helkkula P, Hassan S, Saarentaus E, Vartiainen E, Ruotsalainen S, Leinonen JT, Palotie A, Karjalainen J, Kurki M, Ripatti S, Tukiainen T. Genome-wide association study of varicose veins identifies a protective missense variant in GJD3 enriched in the Finnish population. Commun Biol 2023; 6:71. [PMID: 36653477 PMCID: PMC9849365 DOI: 10.1038/s42003-022-04285-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 11/21/2022] [Indexed: 01/19/2023] Open
Abstract
Varicose veins is the most common manifestation of chronic venous disease that displays female-biased incidence. To identify protein-inactivating variants that could guide identification of drug target genes for varicose veins and genetic evidence for the disease prevalence difference between the sexes, we conducted a genome-wide association study of varicose veins in Finns using the FinnGen dataset with 17,027 cases and 190,028 controls. We identified 50 associated genetic loci (P < 5.0 × 10-8) of which 29 were novel including one near ERG with female-specificity (rs2836405-G, OR[95% CI] = 1.09[1.05-1.13], P = 3.1 × 10-8). These also include two X-chromosomal (ARHGAP6 and SRPX) and two autosomal novel loci (TGFB2 and GJD3) with protein-coding lead variants enriched above 56-fold in Finns over non-Finnish non-Estonian Europeans. A low-frequency missense variant in GJD3 (p.Pro59Thr) is exclusively associated with a lower risk for varicose veins (OR = 0.62 [0.55-0.70], P = 1.0 × 10-14) in a phenome-wide scan of the FinnGen data. The absence of observed pleiotropy and its membership of the connexin gene family underlines GJD3 as a potential connexin-modulating therapeutic strategy for varicose veins. Our results provide insights into varicose veins etiopathology and highlight the power of isolated populations, including Finns, to discover genetic variants that inform therapeutic development.
Collapse
Grants
- MC_PC_17228 Medical Research Council
- Academy of Finland (Suomen Akatemia)
- Sydäntutkimussäätiö (Finnish Foundation for Cardiovascular Research)
- Academy of Finland Center of Excellence in Complex Disease Genetics (Grant No 312062), Sigrid Juselius Foundation (S.Ri. and T.T.), University of Helsinki HiLIFE Fellow and Grand Challenge grants (S.Ri.), University of Helsinki three-year research project grant (T.T.), FIMM-EMBL PhD program doctoral funding (S.H.), Nylands Nation, University of Helsinki (P.H.) The FinnGen project is funded by two grants from Business Finland (HUS 4685/31/2016 and UH 4386/31/2016) and the following industry partners: AbbVie Inc., AstraZeneca UK Ltd, Biogen MA Inc., Bristol Myers Squibb (and Celgene Corporation & Celgene International II Sàrl), Genentech Inc., Merck Sharp & Dohme Corp, Pfizer Inc., GlaxoSmithKline Intellectual Property Development Ltd., Sanofi US Services Inc., Maze Therapeutics Inc., Janssen Biotech Inc, Novartis AG, and Boehringer Ingelheim. Following biobanks are acknowledged for delivering biobank samples to FinnGen: Auria Biobank (www.auria.fi/biopankki), THL Biobank (www.thl.fi/biobank), Helsinki Biobank (www.helsinginbiopankki.fi), Biobank Borealis of Northern Finland (https://www.ppshp.fi/Tutkimus-ja-opetus/Biopankki/Pages/Biobank-Borealis-briefly-in-English.aspx), Finnish Clinical Biobank Tampere (www.tays.fi/en-US/Research_and_development/Finnish_Clinical_Biobank_Tampere), Biobank of Eastern Finland (www.ita-suomenbiopankki.fi/en), Central Finland Biobank (www.ksshp.fi/fi-FI/Potilaalle/Biopankki), Finnish Red Cross Blood Service Biobank (www.veripalvelu.fi/verenluovutus/biopankkitoiminta) and Terveystalo Biobank (www.terveystalo.com/fi/Yritystietoa/Terveystalo-Biopankki/Biopankki/).
Collapse
Affiliation(s)
- Pyry Helkkula
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Shabbeer Hassan
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Elmo Saarentaus
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Emilia Vartiainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Sanni Ruotsalainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Jaakko T Leinonen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Juha Karjalainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Mitja Kurki
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Department of Public Health, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Taru Tukiainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.
| |
Collapse
|
175
|
Dong CX, Malecki C, Robertson E, Hambly B, Jeremy R. Molecular Mechanisms in Genetic Aortopathy-Signaling Pathways and Potential Interventions. Int J Mol Sci 2023; 24:ijms24021795. [PMID: 36675309 PMCID: PMC9865322 DOI: 10.3390/ijms24021795] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Thoracic aortic disease affects people of all ages and the majority of those aged <60 years have an underlying genetic cause. There is presently no effective medical therapy for thoracic aneurysm and surgery remains the principal intervention. Unlike abdominal aortic aneurysm, for which the inflammatory/atherosclerotic pathogenesis is well established, the mechanism of thoracic aneurysm is less understood. This paper examines the key cell signaling systems responsible for the growth and development of the aorta, homeostasis of endothelial and vascular smooth muscle cells and interactions between pathways. The evidence supporting a role for individual signaling pathways in pathogenesis of thoracic aortic aneurysm is examined and potential novel therapeutic approaches are reviewed. Several key signaling pathways, notably TGF-β, WNT, NOTCH, PI3K/AKT and ANGII contribute to growth, proliferation, cell phenotype and survival for both vascular smooth muscle and endothelial cells. There is crosstalk between pathways, and between vascular smooth muscle and endothelial cells, with both synergistic and antagonistic interactions. A common feature of the activation of each is response to injury or abnormal cell stress. Considerable experimental evidence supports a contribution of each of these pathways to aneurysm formation. Although human information is less, there is sufficient data to implicate each pathway in the pathogenesis of human thoracic aneurysm. As some pathways i.e., WNT and NOTCH, play key roles in tissue growth and organogenesis in early life, it is possible that dysregulation of these pathways results in an abnormal aortic architecture even in infancy, thereby setting the stage for aneurysm development in later life. Given the fine tuning of these signaling systems, functional polymorphisms in key signaling elements may set up a future risk of thoracic aneurysm. Multiple novel therapeutic agents have been developed, targeting cell signaling pathways, predominantly in cancer medicine. Future investigations addressing cell specific targeting, reduced toxicity and also less intense treatment effects may hold promise for effective new medical treatments of thoracic aortic aneurysm.
Collapse
Affiliation(s)
- Charlotte Xue Dong
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Cassandra Malecki
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
| | - Elizabeth Robertson
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Brett Hambly
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Richmond Jeremy
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
- Correspondence:
| |
Collapse
|
176
|
Liu M, Xie L, Zhang Y, Chen J, Zhang X, Chen Y, Huang W, Cai M, Liang L, Lai M, Huang J, Guo Y, Lin L, Zhu K. Inhibition of CEMIP potentiates the effect of sorafenib on metastatic hepatocellular carcinoma by reducing the stiffness of lung metastases. Cell Death Dis 2023; 14:25. [PMID: 36639658 PMCID: PMC9839779 DOI: 10.1038/s41419-023-05550-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 12/19/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023]
Abstract
Hepatocellular carcinoma (HCC) with lung metastasis is associated with poor prognosis and poor therapeutic outcomes. Studies have demonstrated that stiffened stroma can promote metastasis in various tumors. However, how the lung mechanical microenvironment favors circulating tumor cells remains unclear in metastatic HCC. Here, we found that the expression of cell migration-inducing hyaluronan-binding protein (CEMIP) was closely associated with lung metastasis and can promote pre-metastatic niche formation by increasing lung matrix stiffness. Furthermore, upregulated serum CEMIP was indicative of lung fibrotic changes severity in patients with HCC lung metastasis. By directly targeting CEMIP, pirfenidone can inhibit CEMIP/TGF-β1/Smad signaling pathway and reduce lung metastases stiffening, demonstrating promising antitumor activity. Pirfenidone in combination with sorafenib can more effectively suppress the incidence of lung metastasis compared with sorafenib alone. This study is the first attempt to modulate the mechanical microenvironment for HCC therapy and highlights CEMIP as a potential target for the prevention and treatment of HCC lung metastasis. CEMIP mediating an HCC-permissive microenvironment through controlling matrix stiffness. Meanwhile, Pirfenidone could reduce metastasis stiffness and increases the anti-angiogenic effect of Sorafenib by directly targeting CEMIP.
Collapse
Affiliation(s)
- Mingyu Liu
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Lulu Xie
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Yuying Zhang
- Central Laboratory, Shenzhen Longhua Maternity and Child Healthcare Hospital, 518109, Shenzhen, China
| | - Jianning Chen
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-Sen University, 510630, Guangzhou, China
| | - Xiang Zhang
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease and The Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ye Chen
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Wensou Huang
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Mingyue Cai
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Licong Liang
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Miaoling Lai
- Department of Pathology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Jingjun Huang
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Yongjian Guo
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China.
| | - Liteng Lin
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China.
| | - Kangshun Zhu
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China.
| |
Collapse
|
177
|
Shanmukha KD, Paluvai H, Lomada SK, Gokara M, Kalangi SK. Histone deacetylase (HDACs) inhibitors: Clinical applications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 198:119-152. [DOI: 10.1016/bs.pmbts.2023.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
178
|
Wang K, Wen D, Xu X, Zhao R, Jiang F, Yuan S, Zhang Y, Gao Y, Li Q. Extracellular matrix stiffness-The central cue for skin fibrosis. Front Mol Biosci 2023; 10:1132353. [PMID: 36968277 PMCID: PMC10031116 DOI: 10.3389/fmolb.2023.1132353] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/20/2023] [Indexed: 03/29/2023] Open
Abstract
Skin fibrosis is a physiopathological process featuring the excessive deposition of extracellular matrix (ECM), which is the main architecture that provides structural support and constitutes the microenvironment for various cellular behaviors. Recently, increasing interest has been drawn to the relationship between the mechanical properties of the ECM and the initiation and modulation of skin fibrosis, with the engagement of a complex network of signaling pathways, the activation of mechanosensitive proteins, and changes in immunoregulation and metabolism. Simultaneous with the progression of skin fibrosis, the stiffness of ECM increases, which in turn perturbs mechanical and humoral homeostasis to drive cell fate toward an outcome that maintains and enhances the fibrosis process, thus forming a pro-fibrotic "positive feedback loop". In this review, we highlighted the central role of the ECM and its dynamic changes at both the molecular and cellular levels in skin fibrosis. We paid special attention to signaling pathways regulated by mechanical cues in ECM remodeling. We also systematically summarized antifibrotic interventions targeting the ECM, hopefully enlightening new strategies for fibrotic diseases.
Collapse
Affiliation(s)
- Kang Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dongsheng Wen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuewen Xu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rui Zhao
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Feipeng Jiang
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Shengqin Yuan
- School of Public Administration, Sichuan University, Chengdu, Sichuan, China
| | - Yifan Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| | - Ya Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| |
Collapse
|
179
|
Itoh F, Watabe T. TGF-β signaling in lymphatic vascular vessel formation and maintenance. Front Physiol 2022; 13:1081376. [PMID: 36589453 PMCID: PMC9799095 DOI: 10.3389/fphys.2022.1081376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Transforming growth factor (TGF)-β and its family members, including bone morphogenetic proteins (BMPs), nodal proteins, and activins, are implicated in the development and maintenance of various organs. Here, we review its role in the lymphatic vascular system (the secondary vascular system in vertebrates), which plays a crucial role in various physiological and pathological processes, participating in the maintenance of the normal tissue fluid balance, immune cell trafficking, and fatty acid absorption in the gut. The lymphatic system is associated with pathogenesis in multiple diseases, including lymphedema, inflammatory diseases, and tumor metastasis. Lymphatic vessels are composed of lymphatic endothelial cells, which differentiate from blood vascular endothelial cells (BECs). Although TGF-β family signaling is essential for maintaining blood vessel function, little is known about the role of TGF-β in lymphatic homeostasis. Recently, we reported that endothelial-specific depletion of TGF-β signaling affects lymphatic function. These reports suggest that TGF-β signaling in lymphatic endothelial cells maintains the structure of lymphatic vessels and lymphatic homeostasis, and promotes tumor lymphatic metastasis. Suppression of TGF-β signaling in lymphatic endothelial cells may therefore be effective in inhibiting cancer metastasis. We highlight recent advances in understanding the roles of TGF-β signaling in the formation and maintenance of the lymphatic system.
Collapse
Affiliation(s)
- Fumiko Itoh
- Laboratory of Stem Cells Regulations, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan,*Correspondence: Fumiko Itoh, ; Tetsuro Watabe,
| | - Tetsuro Watabe
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan,*Correspondence: Fumiko Itoh, ; Tetsuro Watabe,
| |
Collapse
|
180
|
Proteomic Insights into Cardiac Fibrosis: From Pathophysiological Mechanisms to Therapeutic Opportunities. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248784. [PMID: 36557919 PMCID: PMC9781843 DOI: 10.3390/molecules27248784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Cardiac fibrosis is a common pathophysiologic process in nearly all forms of heart disease which refers to excessive deposition of extracellular matrix proteins by cardiac fibroblasts. Activated fibroblasts are the central cellular effectors in cardiac fibrosis, and fibrotic remodelling can cause several cardiac dysfunctions either by reducing the ejection fraction due to a stiffened myocardial matrix, or by impairing electric conductance. Recently, there is a rising focus on the proteomic studies of cardiac fibrosis for pathogenesis elucidation and potential biomarker mining. This paper summarizes the current knowledge of molecular mechanisms underlying cardiac fibrosis, discusses the potential of imaging and circulating biomarkers available to recognize different phenotypes of this lesion, reviews the currently available and potential future therapies that allow individualized management in reversing progressive fibrosis, as well as the recent progress on proteomic studies of cardiac fibrosis. Proteomic approaches using clinical specimens and animal models can provide the ability to track pathological changes and new insights into the mechanisms underlining cardiac fibrosis. Furthermore, spatial and cell-type resolved quantitative proteomic analysis may also serve as a minimally invasive method for diagnosing cardiac fibrosis and allowing for the initiation of prophylactic treatment.
Collapse
|
181
|
Spira A, Awada A, Isambert N, Lorente D, Penel N, Zhang Y, Ojalvo LS, Hicking C, Rolfe PA, Ihling C, Dussault I, Locke G, Borel C. Identification of HMGA2 as a predictive biomarker of response to bintrafusp alfa in a phase 1 trial in patients with advanced triple-negative breast cancer. Front Oncol 2022; 12:981940. [PMID: 36568239 PMCID: PMC9773992 DOI: 10.3389/fonc.2022.981940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/28/2022] [Indexed: 12/13/2022] Open
Abstract
Background We report the clinical activity, safety, and identification of a predictive biomarker for bintrafusp alfa, a first-in-class bifunctional fusion protein composed of the extracellular domain of TGFβRII (a TGF-β "trap") fused to a human IgG1 mAb blocking PD-L1, in patients with advanced triple-negative breast cancer (TNBC). Methods In this expansion cohort of a global phase 1 study, patients with pretreated, advanced TNBC received bintrafusp alfa 1200 mg every 2 weeks intravenously until disease progression, unacceptable toxicity, or withdrawal. The primary objective was confirmed best overall response by RECIST 1.1 assessed per independent review committee (IRC). Results As of May 15, 2020, a total of 33 patients had received bintrafusp alfa, for a median of 6.0 (range, 2.0-48.1) weeks. The objective response rate was 9.1% (95% CI, 1.9%-24.3%) by IRC and investigator assessment. The median progression-free survival per IRC was 1.3 (95% CI, 1.2-1.4) months, and median overall survival was 7.7 (95% CI, 2.1-10.9) months. Twenty-five patients (75.8%) experienced treatment-related adverse events (TRAEs). Grade 3 TRAEs occurred in 5 patients (15.2%); no patients had a grade 4 TRAE. There was 1 treatment-related death (dyspnea, hemolysis, and thrombocytopenia in a patient with extensive disease at trial entry). Responses occurred independently of PD-L1 expression, and tumor RNAseq data identified HMGA2 as a potential biomarker of response. Conclusions Bintrafusp alfa showed clinical activity and manageable safety in patients with heavily pretreated advanced TNBC. HMGA2 was identified as a potential predictive biomarker of response. ClinicalTrialsgov identifier NCT02517398.
Collapse
Affiliation(s)
- Alexander Spira
- Department of Medical Oncology, Virginia Cancer Specialists, Fairfax, VA, United States
- US Oncology Research, The Woodlands, TX, United States
| | - Ahmad Awada
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Nicolas Isambert
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | - David Lorente
- Department of Medical Oncology, Hospital Universitari I Politècnic La Fe, Valencia, Spain
| | - Nicolas Penel
- Department of Medical Oncology, Centre Oscar Lambret, Lille, France
- Department of Medical Oncology, Université de Lille, Lille, France
| | - Yue Zhang
- EMD Serono Research & Development Institute, Inc, an Affiliate of Merck KGaA, Billerica, MA, United States
| | - Laureen S. Ojalvo
- EMD Serono Research & Development Institute, Inc, an Affiliate of Merck KGaA, Billerica, MA, United States
| | | | - P. Alexander Rolfe
- EMD Serono Research & Development Institute, Inc, an Affiliate of Merck KGaA, Billerica, MA, United States
| | | | - Isabelle Dussault
- EMD Serono Research & Development Institute, Inc, an Affiliate of Merck KGaA, Billerica, MA, United States
| | - George Locke
- EMD Serono Research & Development Institute, Inc, an Affiliate of Merck KGaA, Billerica, MA, United States
| | - Christian Borel
- Department of Medical Oncology, Centre Paul Strauss, Strasbourg, France
| |
Collapse
|
182
|
Agashe RP, Lippman SM, Kurzrock R. JAK: Not Just Another Kinase. Mol Cancer Ther 2022; 21:1757-1764. [PMID: 36252553 PMCID: PMC10441554 DOI: 10.1158/1535-7163.mct-22-0323] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/07/2022] [Accepted: 10/07/2022] [Indexed: 01/12/2023]
Abstract
The JAK/STAT axis is implicated in cancer, inflammation, and immunity. Numerous cytokines/growth factors affect JAK/STAT signaling. JAKs (JAK1, JAK2, JAK3, and TYK2) noncovalently associate with cytokine receptors, mediate receptor tyrosine phosphorylation, and recruit ≥1 STAT proteins (STAT1, STAT2, STAT3, STAT4, STAT5a, STAT5b, and STAT6). Tyrosine-phosphorylated STATs dimerize and are then transported into the nucleus to function as transcription factors. Signaling is attenuated by specific suppressor of cytokine signaling proteins, creating a negative feedback loop. Both germline mutations and polymorphisms of JAK family members correlate with specific diseases: Systemic lupus erythematosus (TYK2 polymorphisms); severe combined immunodeficiency (JAK3 mutations); pediatric acute lymphoblastic leukemia (TYK2 mutations); and hereditary thrombocytosis (JAK2 mutations). Somatic gain-of-function JAK mutations mainly occur in hematologic malignancies, with the activating JAK2 V617F being a myeloproliferative disorder hallmark; it is also seen in clonal hematopoiesis of indeterminate potential. Several T-cell malignancies, as well as B-cell acute lymphoblastic leukemia, and acute megakaryoblastic leukemia also harbor JAK family somatic alterations. On the other hand, JAK2 copy-number loss is associated with immune checkpoint inhibitor resistance. JAK inhibitors (jakinibs) have been deployed in many conditions with JAK activation; they are approved in myeloproliferative disorders, rheumatoid and psoriatic arthritis, atopic dermatitis, ulcerative colitis, graft-versus-host disease, alopecia areata, ankylosing spondylitis, and in patients hospitalized for COVID-19. Clinical trials are investigating jakinibs in multiple other autoimmune/inflammatory conditions. Furthermore, dermatologic and neurologic improvements have been observed in children with Aicardi-Goutieres syndrome (a genetic interferonopathy) treated with JAK inhibitors.
Collapse
Affiliation(s)
| | | | - Razelle Kurzrock
- Medical College of Wisconsin, Milwaukee, Wisconsin
- Win Consortium, Paris, France
| |
Collapse
|
183
|
Cobb MS, Tao S, Shortt K, Girgis M, Hauptman J, Schriewer J, Chin Z, Dorfman E, Campbell K, Heruth DP, Shohet RV, Dawn B, Konorev EA. Smad3 promotes adverse cardiovascular remodeling and dysfunction in doxorubicin-treated hearts. Am J Physiol Heart Circ Physiol 2022; 323:H1091-H1107. [PMID: 36269647 PMCID: PMC9678413 DOI: 10.1152/ajpheart.00312.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/26/2022] [Accepted: 10/14/2022] [Indexed: 01/21/2023]
Abstract
Many anticancer therapies cause serious cardiovascular complications that degrade quality of life and cause early mortality in treated patients. Specifically, doxorubicin is known as an effective anticancer agent that causes cardiomyopathy in treated patients. There has been growing interest in defining the role of endothelial cells in cardiac damage by doxorubicin. We have shown in the present study that endothelial nuclei accumulate more intravenously administered doxorubicin than other cardiac cell types. Doxorubicin enhanced cardiac production of the transforming growth factor-β (TGF-β) ligands and nuclear translocation of phospho-Smad3 in both cultured and in vivo cardiac endothelial cells. To examine the role of the TGF-β/mothers against decapentaplegic homolog 3 (Smad3) pathway in cardiac damage by doxorubicin, we used both Smad3 shRNA stable endothelial cell lines and Smad3-knockout mice. We demonstrated using endothelial transcriptome analysis that upregulation of the TGF-β and inflammatory cytokine/cytokine receptor pathways, as well as suppression of cell cycle and angiogenesis by doxorubicin, were alleviated in Smad3-deficient endothelial cells. The results of transcriptomic analysis were validated using qPCR, immunoblotting, and ex vivo aortic ring sprouting assays. Similarly, increased cardiac expression of cytokines and chemokines observed in treated wild-type mice was diminished in treated Smad3-knockout animals. We also detected increased end-diastolic diameter and depressed systolic function in doxorubicin-treated wild-type but not Smad3-knockout mice. This work provides evidence for the critical role of the canonical TGF-β/Smad3 pathway in cardiac damage by doxorubicin.NEW & NOTEWORTHY Microvascular endothelial cells in the heart accumulate more intravenously administered doxorubicin than nonendothelial cardiac cell types. The treatment enhanced the TGF-β/Smad3 pathway and elicited endothelial cell senescence and inflammatory responses followed by adverse cardiac remodeling and dysfunction in wild-type but not Smad3-deficient animals. Our study suggests that the TGF-β/Smad3 pathway contributes to the development of doxorubicin cardiomyopathy and the potential value of novel approaches to ameliorate cardiotoxicity by targeting the Smad3 transcription factor.
Collapse
Affiliation(s)
- Melissa S Cobb
- Department of Basic Sciences, Kansas City University, Kansas City, Missouri
| | - Shixin Tao
- Department of Basic Sciences, Kansas City University, Kansas City, Missouri
| | - Katherine Shortt
- Ambry Genetics, Department of Advanced Analytics, Aliso Viejo, California
| | - Magdy Girgis
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at UNLV, Las Vegas, Nevada
| | - Jeryl Hauptman
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at UNLV, Las Vegas, Nevada
| | - Jill Schriewer
- Department of Basic Sciences, Kansas City University, Kansas City, Missouri
| | - Zaphrirah Chin
- Department of Basic Sciences, Kansas City University, Kansas City, Missouri
| | - Edward Dorfman
- Department of Basic Sciences, Kansas City University, Kansas City, Missouri
| | - Kyle Campbell
- Department of Basic Sciences, Kansas City University, Kansas City, Missouri
| | - Daniel P Heruth
- The Children's Mercy Research Institute, Kansas City, Missouri
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Ralph V Shohet
- Department of Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Buddhadeb Dawn
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at UNLV, Las Vegas, Nevada
| | - Eugene A Konorev
- Department of Basic Sciences, Kansas City University, Kansas City, Missouri
| |
Collapse
|
184
|
Wang Y, Mack JA, Hascall VC, Maytin EV. Transforming Growth Factor-β Receptor-Mediated, p38 Mitogen-Activated Protein Kinase-Dependent Signaling Drives Enhanced Myofibroblast Differentiation during Skin Wound Healing in Mice Lacking Hyaluronan Synthases 1 and 3. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1683-1698. [PMID: 36063901 PMCID: PMC9765314 DOI: 10.1016/j.ajpath.2022.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 06/30/2022] [Accepted: 08/15/2022] [Indexed: 12/31/2022]
Abstract
Normal myofibroblast differentiation is critical for proper skin wound healing. Neoexpression of α-smooth muscle actin (α-SMA), a marker for myofibroblast differentiation, is driven by transforming growth factor (TGF)-β receptor-mediated signaling. Hyaluronan and its three synthesizing enzymes, hyaluronan synthases (Has 1, 2, and 3), also participate in this process. Closure of skin wounds is significantly accelerated in Has1/3 double-knockout (Has1/3-null) mice. Herein, TGF-β activity and dermal collagen maturation were increased in Has1/3-null healing skin. Cultures of primary skin fibroblasts isolated from Has1/3-null mice had higher levels of TGF-β activity, α-SMA expression, and phosphorylation of p38 mitogen-activated protein kinase at Thr180/Tyr182, compared with wild-type fibroblasts. p38α mitogen-activated protein kinase was a necessary element in a noncanonical TGF-β receptor signaling pathway driving α-SMA expression in Has1/3-null fibroblasts. Myocardin-related transcription factor (MRTF), a cofactor that binds to the transcription factor serum response factor (SRF), was also critical. Nuclear localization of MRTF was increased, and MRTF binding to SRF was enhanced in Has1/3-null fibroblasts. Inhibition of MRTF or SRF expression by RNA interference suppresses α-SMA expression at baseline and diminished its overexpression in Has1/3-null fibroblasts. Interestingly, total matrix metalloproteinase activity was increased in healing skin and fibroblasts from Has1/3-null mice, possibly explaining the increased TGF-β activation.
Collapse
Affiliation(s)
- Yan Wang
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio.
| | - Judith A Mack
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio; Department of Dermatology, Dermatology and Plastic Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| | - Vincent C Hascall
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Edward V Maytin
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio; Department of Dermatology, Dermatology and Plastic Surgery Institute, Cleveland Clinic, Cleveland, Ohio.
| |
Collapse
|
185
|
Lo Iacono M, Gaggianesi M, Bianca P, Brancato OR, Muratore G, Modica C, Roozafzay N, Shams K, Colarossi L, Colarossi C, Memeo L, Turdo A, Veschi V, Di Franco S, Todaro M, Stassi G. Destroying the Shield of Cancer Stem Cells: Natural Compounds as Promising Players in Cancer Therapy. J Clin Med 2022; 11:6996. [PMID: 36498571 PMCID: PMC9737492 DOI: 10.3390/jcm11236996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
In a scenario where eco-sustainability and a reduction in chemotherapeutic drug waste are certainly a prerogative to safeguard the biosphere, the use of natural products (NPs) represents an alternative therapeutic approach to counteract cancer diseases. The presence of a heterogeneous cancer stem cell (CSC) population within a tumor bulk is related to disease recurrence and therapy resistance. For this reason, CSC targeting presents a promising strategy for hampering cancer recurrence. Increasing evidence shows that NPs can inhibit crucial signaling pathways involved in the maintenance of CSC stemness and sensitize CSCs to standard chemotherapeutic treatments. Moreover, their limited toxicity and low costs for large-scale production could accelerate the use of NPs in clinical settings. In this review, we will summarize the most relevant studies regarding the effects of NPs derived from major natural sources, e.g., food, botanical, and marine species, on CSCs, elucidating their use in pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Melania Lo Iacono
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Miriam Gaggianesi
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Paola Bianca
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Ornella Roberta Brancato
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Giampaolo Muratore
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Chiara Modica
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Narges Roozafzay
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Kimiya Shams
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Lorenzo Colarossi
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, 95029 Catania, Italy
| | - Cristina Colarossi
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, 95029 Catania, Italy
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, 95029 Catania, Italy
| | - Alice Turdo
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Veronica Veschi
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Simone Di Franco
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Matilde Todaro
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Giorgio Stassi
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
186
|
Ye Z, Kilic G, Dabelsteen S, Marinova IN, Thøfner JF, Song M, Rudjord-Levann AM, Bagdonaite I, Vakhrushev SY, Brakebusch CH, Olsen JV, Wandall HH. Characterization of TGF-β signaling in a human organotypic skin model reveals that loss of TGF-βRII induces invasive tissue growth. Sci Signal 2022; 15:eabo2206. [DOI: 10.1126/scisignal.abo2206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Transforming growth factor–β (TGF-β) signaling regulates various aspects of cell growth and differentiation and is often dysregulated in human cancers. We combined genetic engineering of a human organotypic three-dimensional (3D) skin model with global quantitative proteomics and phosphoproteomics to dissect the importance of essential components of the TGF-β signaling pathway, including the ligands TGF-β1, TGF-β2, and TGF-β3, the receptor TGF-βRII, and the intracellular effector SMAD4. Consistent with the antiproliferative effects of TGF-β signaling, the loss of TGF-β1 or SMAD4 promoted cell cycling and delayed epidermal differentiation. The loss of TGF-βRII, which abrogates both SMAD4-dependent and SMAD4-independent downstream signaling, more strongly affected cell proliferation and differentiation than did loss of SMAD4, and it induced invasive growth. TGF-βRII knockout reduced cell-matrix interactions, and the production of matrix proteins increased the production of cancer-associated cell-cell adhesion proteins and proinflammatory mediators and increased mitogen-activated protein kinase (MAPK) signaling. Inhibiting the activation of the ERK and p38 MAPK pathways blocked the development of the invasive phenotype upon the loss of TGF-βRII. This study provides a framework for exploring TGF-β signaling pathways in human epithelial tissue homeostasis and transformation using genetic engineering, 3D tissue models, and high-throughput quantitative proteomics and phosphoproteomics.
Collapse
Affiliation(s)
- Zilu Ye
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gülcan Kilic
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Section of Oral Biology and Immunopathology, School of Dentistry, University of Copenhagen, Copenhagen, Denmark
| | - Sally Dabelsteen
- Section of Oral Biology and Immunopathology, School of Dentistry, University of Copenhagen, Copenhagen, Denmark
| | - Irina N. Marinova
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens F. B. Thøfner
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ming Song
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Asha M. Rudjord-Levann
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ieva Bagdonaite
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sergey Y. Vakhrushev
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cord H. Brakebusch
- Biotech Research and Innovation Centre, Biomedical Institute, University of Copenhagen, Copenhagen, Denmark
| | - Jesper V. Olsen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans H. Wandall
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
187
|
Li X, Wu Y, Tian T. TGF-β Signaling in Metastatic Colorectal Cancer (mCRC): From Underlying Mechanism to Potential Applications in Clinical Development. Int J Mol Sci 2022; 23:14436. [PMID: 36430910 PMCID: PMC9698504 DOI: 10.3390/ijms232214436] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/08/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Colorectal cancer (CRC) is a serious public health issue, and it has the leading incidence and mortality among malignant tumors worldwide. CRC patients with metastasis in the liver, lung or other distant sites always have poor prognosis. Thus, there is an urgent need to discover the underlying mechanisms of metastatic colorectal cancer (mCRC) and to develop optimal therapy for mCRC. Transforming growth factor-β (TGF-β) signaling plays a significant role in various physiologic and pathologic processes, and aberrant TGF-β signal transduction contributes to mCRC progression. In this review, we summarize the alterations of the TGF-β signaling pathway in mCRC patients, the functional mechanisms of TGF-β signaling, its promotion of epithelial-mesenchymal transition, its facilitation of angiogenesis, its suppression of anti-tumor activity of immune cells in the microenvironment and its contribution to stemness of CRC cells. We also discuss the possible applications of TGF-β signaling in mCRC diagnosis, prognosis and targeted therapies in clinical trials. Hopefully, these research advances in TGF-β signaling in mCRC will improve the development of new strategies that can be combined with molecular targeted therapy, immunotherapy and traditional therapies to achieve better efficacy and benefit mCRC patients in the near future.
Collapse
Affiliation(s)
| | | | - Tian Tian
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| |
Collapse
|
188
|
D'Alessandro VF, Takeshita A, Yasuma T, Toda M, D'Alessandro-Gabazza CN, Okano Y, Tharavecharak S, Inoue C, Nishihama K, Fujimoto H, Kobayashi T, Yano Y, Gabazza EC. Transforming Growth Factorβ1 Overexpression Is Associated with Insulin Resistance and Rapidly Progressive Kidney Fibrosis under Diabetic Conditions. Int J Mol Sci 2022; 23:ijms232214265. [PMID: 36430743 PMCID: PMC9693927 DOI: 10.3390/ijms232214265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/08/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
Diabetes mellitus is a global health problem. Diabetic nephropathy is a common complication of diabetes mellitus and the leading cause of end-stage renal disease. The clinical course, response to therapy, and prognosis of nephropathy are worse in diabetic than in non-diabetic patients. The role of transforming growth factorβ1 in kidney fibrosis is undebatable. This study assessed whether the overexpression of transforming growth factorβ1 is associated with insulin resistance and the rapid progression of transforming growth factorβ1-mediated nephropathy under diabetic conditions. Diabetes mellitus was induced with streptozotocin in wild-type mice and transgenic mice with the kidney-specific overexpression of human transforming growth factorβ1. Mice treated with saline were the controls. Glucose tolerance and kidney fibrosis were evaluated. The blood glucose levels, the values of the homeostasis model assessment for insulin resistance, and the area of kidney fibrosis were significantly increased, and the renal function was significantly impaired in the diabetic transforming growth factorβ1 transgenic mice compared to the non-diabetic transgenic mice, diabetic wild-type mice, and non-diabetic mice. Transforming growth factorβ1 impaired the regulatory effect of insulin on glucose in the hepatocyte and skeletal muscle cell lines. This study shows that transforming growth factorβ1 overexpression is associated with insulin resistance and rapidly progressive kidney fibrosis under diabetic conditions in mice.
Collapse
Affiliation(s)
- Valeria Fridman D'Alessandro
- Department of Immunology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Atsuro Takeshita
- Department of Immunology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
- Department of Diabetes and Endocrinology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Taro Yasuma
- Department of Immunology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
- Department of Diabetes and Endocrinology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Masaaki Toda
- Department of Immunology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Corina N D'Alessandro-Gabazza
- Department of Immunology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Yuko Okano
- Department of Immunology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
- Department of Diabetes and Endocrinology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Suphachai Tharavecharak
- Department of Immunology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Chisa Inoue
- Department of Diabetes and Endocrinology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Kota Nishihama
- Department of Diabetes and Endocrinology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Hajime Fujimoto
- Department of Pulmonary and Critical care Medicine, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Tetsu Kobayashi
- Department of Pulmonary and Critical care Medicine, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Yutaka Yano
- Department of Diabetes and Endocrinology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Esteban C Gabazza
- Department of Immunology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| |
Collapse
|
189
|
Zhou Q, Yuan O, Cui H, Hu T, Xiao GG, Wei J, Zhang H, Wu C. Bioinformatic analysis identifies HPV-related tumor microenvironment remodeling prognostic biomarkers in head and neck squamous cell carcinoma. Front Cell Infect Microbiol 2022; 12:1007950. [PMID: 36425786 PMCID: PMC9679011 DOI: 10.3389/fcimb.2022.1007950] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 10/10/2022] [Indexed: 08/29/2023] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are highly aggressive tumors with rapid progression and poor prognosis. Human papillomavirus (HPV) infection has been identified as one of the most important carcinogens for HNSCC. As an early event in HNSCC, infection with HPV leads to altered immune profiles in the tumor microenvironment (TME). The TME plays a key role in the progression and transformation of HNSCC. However, the TME in HNSCC is a complex and heterogeneous mix of tumor cells, fibroblasts, different types of infiltrating immune cells, and extracellular matrix. Biomarkers relevant to the TME, and the biological role of these biomarkers, remain poorly understood. To this end, we performed comprehensive analysis of the RNA sequencing (RNA-Seq) data from tumor tissue of 502 patients with HNSCC and healthy tissue of 44 control samples. In total, we identified 4,237 differentially expressed genes, including 2,062 upregulated and 2,175 downregulated genes. Further in-depth bioinformatic analysis suggested 19 HNSCC tumor tissue-specific genes. In the subsequent analysis, we focused on the biomarker candidates shown to be significantly associated with unfavorable patient survival: ITGA5, PLAU, PLAUR, SERPINE1, TGFB1, and VEGFC. We found that the expression of these genes was negatively regulated by DNA methylation. Strikingly, all of these potential biomarkers are profoundly involved in the activation of the epithelial-mesenchymal transition (EMT) pathway in HNSCCs. In addition, these targets were found to be positively correlated with the immune invasion levels of CD4+ T cells, macrophages, neutrophils, and dendritic cells, but negatively correlated with B-cell infiltration and CD8+ T-cell invasion. Notably, our data showed that the expression levels of ITGA5, PLAU, PLAUR, SERPINE1, and TGFB1 were significantly overexpressed in HPV-positive HNSCCs compared to normal controls, indicating the potential role of these biomarkers as transformation and/or malignant progression markers for HNSCCs in patients with HPV infection. Taken together, the results of our study propose ITGA5, PLAU, PLAUR, SERPINE1, and TGFB1 as potential prognostic biomarkers for HNSCCs, which might be involved in the HPV-related TME remodeling of HNSCC. Our findings provide important implications for the development and/or improvement of patient stratification and customized immunotherapies in HNSCC.
Collapse
Affiliation(s)
- Qimin Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ouyang Yuan
- Department of Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongtu Cui
- School of Biomedical Engineering, Dalian University of Technology, Dalian, China
| | - Tao Hu
- School of Biomedical Engineering, Dalian University of Technology, Dalian, China
| | - Gary Guishan Xiao
- School of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China
| | - Jiao Wei
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Honglei Zhang
- Department of Otolaryngology Head and Neck Surgery, Air Force Medical Centre, People's Liberation Army (PLA), Beijing, China
| | - Chengjun Wu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
190
|
Quach HT, Hou Z, Bellis RY, Saini JK, Amador-Molina A, Adusumilli PS, Xiong Y. Next-generation immunotherapy for solid tumors: combination immunotherapy with crosstalk blockade of TGFβ and PD-1/PD-L1. Expert Opin Investig Drugs 2022; 31:1187-1202. [PMID: 36448335 PMCID: PMC10085570 DOI: 10.1080/13543784.2022.2152323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022]
Abstract
INTRODUCTION In solid tumor immunotherapy, less than 20% of patients respond to anti-programmed cell death 1 (PD-1)/programmed cell death 1 ligand 1 (PD-L1) agents. The role of transforming growth factor β (TGFβ) in diverse immunity is well-established; however, systemic blockade of TGFβ is associated with toxicity. Accumulating evidence suggests the role of crosstalk between TGFβ and PD-1/PD-L1 pathways. AREAS COVERED We focus on TGFβ and PD-1/PD-L1 signaling pathway crosstalk and the determinant role of TGFβ in the resistance of immune checkpoint blockade. We provide the rationale for combination anti-TGFβ and anti-PD-1/PD-L1 therapies for solid tumors and discuss the current status of dual blockade therapy in preclinical and clinical studies. EXPERT OPINION The heterogeneity of tumor microenvironment across solid tumors complicates patient selection, treatment regimens, and response and toxicity assessment for investigation of dual blockade agents. However, clinical knowledge from single-agent studies provides infrastructure to translate dual blockade therapies. Dual TGFβ and PD-1/PD-L1 blockade results in enhanced T-cell infiltration into tumors, a primary requisite for successful immunotherapy. A bifunctional fusion protein specifically targets TGFβ in the tumor microenvironment, avoiding systemic toxicity, and prevents interaction of PD-1+ cytotoxic cells with PD-L1+ tumor cells.
Collapse
Affiliation(s)
- Hue Tu Quach
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Zhaohua Hou
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Rebecca Y. Bellis
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Jasmeen K. Saini
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Alfredo Amador-Molina
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Prasad S. Adusumilli
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Director, Mesothelioma Program; Head, Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Yuquan Xiong
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| |
Collapse
|
191
|
Ruan Q, Lin X, Wang L, Wang N, Zhao Y, Wang H, Tian FY, Hu N, Li Y, Zhao B. An engineered (CAGA)12-EGFP cell-based biosensor for high-content and accurate detection of active TGF-β. Biosens Bioelectron 2022; 220:114884. [DOI: 10.1016/j.bios.2022.114884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/10/2022]
|
192
|
Barcellos-Hoff MH. The radiobiology of TGFβ. Semin Cancer Biol 2022; 86:857-867. [PMID: 35122974 DOI: 10.1016/j.semcancer.2022.02.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 01/27/2023]
Abstract
Ionizing radiation is a pillar of cancer therapy that is deployed in more than half of all malignancies. The therapeutic effect of radiation is attributed to induction of DNA damage that kills cancers cells, but radiation also affects signaling that alters the composition of the tumor microenvironment by activating transforming growth factor β (TGFβ). TGFβ is a ubiquitously expressed cytokine that acts as biological lynchpin to orchestrate phenotypes, the stroma, and immunity in normal tissue; these activities are subverted in cancer to promote malignancy, a permissive tumor microenvironment and immune evasion. The radiobiology of TGFβ unites targets at the forefront of oncology-the DNA damage response and immunotherapy. The cancer cell intrinsic and extrinsic network of TGFβ responses in the irradiated tumor form a barrier to both genotoxic treatments and immunotherapy response. Here, we focus on the mechanisms by which radiation induces TGFβ activation, how TGFβ regulates DNA repair, and the dynamic regulation of the tumor immune microenvironment that together oppose effective cancer therapy. Strategies to inhibit TGFβ exploit fundamental radiobiology that may be the missing link to deploying TGFβ inhibitors for optimal patient benefit from cancer treatment.
Collapse
Affiliation(s)
- Mary Helen Barcellos-Hoff
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
193
|
Yao Z, Boyce BF. TGFβ priming promotes TNF-induced bone erosion: a promising new target in RA? Nat Rev Rheumatol 2022; 18:617-618. [PMID: 36114427 DOI: 10.1038/s41584-022-00843-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Zhenqiang Yao
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Brendan F Boyce
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
194
|
Pujala B, Ramachandran SA, Sonawane M, Kamble MM, Panpatil D, Adhikari S, Soni S, Subbareddy V, Shinde BU, Nayak AK, Bansal C, Gupta A, Mukherjee K, Agarwal AK, Guerrero J, Herrera FJ, Bernales S, Guha M, Chakravarty S, Pham SM, Rai R. Discovery of MDV6058 (PF-06952229), a selective and potent TGFβR1 inhibitor: Design, synthesis and optimization. Bioorg Med Chem Lett 2022; 75:128979. [PMID: 36089110 DOI: 10.1016/j.bmcl.2022.128979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/25/2022] [Accepted: 09/04/2022] [Indexed: 11/15/2022]
Abstract
Compound 1 is a potent TGF-β receptor type-1 (TGFβR1 or ALK5) inhibitor but is metabolically unstable. A solvent-exposed part of this molecule was used to analogue and modulate cell activity, liver microsome stability and mouse pharmacokinetics. The evolution of SAR that led to the selection of 2 (MDV6058 / PF-06952229) as a preclinical lead compound is described.
Collapse
Affiliation(s)
- Brahmam Pujala
- Integral BioSciences Pvt. Ltd., C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Sreekanth A Ramachandran
- Integral BioSciences Pvt. Ltd., C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Mukesh Sonawane
- Integral BioSciences Pvt. Ltd., C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Maruti M Kamble
- Integral BioSciences Pvt. Ltd., C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Dayanand Panpatil
- Integral BioSciences Pvt. Ltd., C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Swati Adhikari
- Integral BioSciences Pvt. Ltd., C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Sanjeev Soni
- Integral BioSciences Pvt. Ltd., C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Venkata Subbareddy
- Integral BioSciences Pvt. Ltd., C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Bharat U Shinde
- Integral BioSciences Pvt. Ltd., C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Anjan K Nayak
- Integral BioSciences Pvt. Ltd., C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Chandni Bansal
- Integral BioSciences Pvt. Ltd., C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Ashu Gupta
- Integral BioSciences Pvt. Ltd., C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Kakoli Mukherjee
- Integral BioSciences Pvt. Ltd., C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Anil K Agarwal
- Integral BioSciences Pvt. Ltd., C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Javier Guerrero
- Fundación Ciencia & Vida, Avenida Zañartu 1482, Ñuñoa, Santiago, 778027, Chile
| | - Francisco J Herrera
- Medivation, Inc., 525 Market Street, 36(th) Floor, San Francisco, CA 94105, USA
| | - Sebastian Bernales
- Medivation, Inc., 525 Market Street, 36(th) Floor, San Francisco, CA 94105, USA
| | - Mausumee Guha
- Medivation, Inc., 525 Market Street, 36(th) Floor, San Francisco, CA 94105, USA; Pfizer Inc., Pfizer World Wide Research & Development, Building CB-4, 10777 Science Center Dr. La Jolla, CA 92121-1111, USA
| | | | - Son M Pham
- Medivation, Inc., 525 Market Street, 36(th) Floor, San Francisco, CA 94105, USA
| | - Roopa Rai
- Medivation, Inc., 525 Market Street, 36(th) Floor, San Francisco, CA 94105, USA.
| |
Collapse
|
195
|
Immunoregulatory signal networks and tumor immune evasion mechanisms: insights into therapeutic targets and agents in clinical development. Biochem J 2022; 479:2219-2260. [DOI: 10.1042/bcj20210233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/17/2022]
Abstract
Through activation of immune cells, the immune system is responsible for identifying and destroying infected or otherwise damaged cells including tumorigenic cells that can be recognized as foreign, thus maintaining homeostasis. However, tumor cells have evolved several mechanisms to avoid immune cell detection and killing, resulting in tumor growth and progression. In the tumor microenvironment, tumor infiltrating immune cells are inactivated by soluble factors or tumor promoting conditions and lose their effects on tumor cells. Analysis of signaling and crosstalk between immune cells and tumor cells have helped us to understand in more detail the mechanisms of tumor immune evasion and this forms basis for drug development strategies in the area of cancer immunotherapy. In this review, we will summarize the dominant signaling networks involved in immune escape and describe the status of development of therapeutic strategies to target tumor immune evasion mechanisms with focus on how the tumor microenvironment interacts with T cells.
Collapse
|
196
|
Moittié S, Jarvis R, Bandelow S, Byrne S, Dobbs P, Grant M, Reeves C, White K, Liptovszky M, Baiker K. Vitamin D status in chimpanzees in human care: a Europe wide study. Sci Rep 2022; 12:17625. [PMID: 36271125 PMCID: PMC9587231 DOI: 10.1038/s41598-022-21211-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 09/23/2022] [Indexed: 01/13/2023] Open
Abstract
While vitamin D deficiency is a public health concern in humans, comparatively little is known about vitamin D levels in non-human primates. Vitamin D plays a crucial role in overall health and its deficiency is associated with a range of disorders, including cardiovascular disease, which is a leading cause of death in great apes. Serum samples (n = 245) from chimpanzees (Pan troglodytes) housed at 32 European zoos were measured for 25-hydroxyvitamin D2, 25-hydroxyvitamin D3 and total 25-hydroxyvitamin D (25-OHD) using liquid chromatography and tandem mass spectrometry. Of these samples, 33.1% indicated inadequate vitamin D status, using the human reference interval (25-OHD < 50 nmol/L). The season of the year, health status of the animal, and the provision of daily outdoor access had a significant effect on vitamin D status. This is the first large-scale study on vitamin D status of non-human great apes in human care. Inadequate 25-OHD serum concentrations are widespread in the chimpanzee population in Europe and could be a risk factor for the development of idiopathic myocardial fibrosis, a major cause of mortality in this species, as well as other diseases. A review of husbandry and nutrition practices is recommended to ensure optimal vitamin D supply for these endangered animals.
Collapse
Affiliation(s)
- Sophie Moittié
- School of Veterinary Medicine, St. George's University, West Indies, Grenada.
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, LE12 5RD, UK.
| | - Rachel Jarvis
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, LE12 5RD, UK.
| | - Stephan Bandelow
- School of Medicine, St. George's University, West Indies, Grenada
| | - Sarah Byrne
- Twycross Zoo, Atherstone, CV9 3PX, UK
- Dublin Zoo, Saint James', Dublin 8, Ireland
| | | | - Melissa Grant
- School of Dentistry, Institute of Clinical Science, University of Birmingham and Birmingham Community Healthcare Foundation Trust, Birmingham, B5 7ET, UK
| | - Christopher Reeves
- Department of Clinical Biochemistry, Manchester University NHS Foundation Trust, Manchester, UK
| | - Kate White
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, LE12 5RD, UK
| | - Mátyás Liptovszky
- Twycross Zoo, Atherstone, CV9 3PX, UK
- Department of Life Sciences, Perth Zoo, South Perth, WA, 6151, Australia
| | - Kerstin Baiker
- Department of Veterinary Clinical Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, Hong Kong
| |
Collapse
|
197
|
Tiwari A, Trivedi R, Lin SY. Tumor microenvironment: barrier or opportunity towards effective cancer therapy. J Biomed Sci 2022; 29:83. [PMID: 36253762 PMCID: PMC9575280 DOI: 10.1186/s12929-022-00866-3] [Citation(s) in RCA: 166] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 10/01/2022] [Indexed: 12/24/2022] Open
Abstract
Tumor microenvironment (TME) is a specialized ecosystem of host components, designed by tumor cells for successful development and metastasis of tumor. With the advent of 3D culture and advanced bioinformatic methodologies, it is now possible to study TME’s individual components and their interplay at higher resolution. Deeper understanding of the immune cell’s diversity, stromal constituents, repertoire profiling, neoantigen prediction of TMEs has provided the opportunity to explore the spatial and temporal regulation of immune therapeutic interventions. The variation of TME composition among patients plays an important role in determining responders and non-responders towards cancer immunotherapy. Therefore, there could be a possibility of reprogramming of TME components to overcome the widely prevailing issue of immunotherapeutic resistance. The focus of the present review is to understand the complexity of TME and comprehending future perspective of its components as potential therapeutic targets. The later part of the review describes the sophisticated 3D models emerging as valuable means to study TME components and an extensive account of advanced bioinformatic tools to profile TME components and predict neoantigens. Overall, this review provides a comprehensive account of the current knowledge available to target TME.
Collapse
Affiliation(s)
- Aadhya Tiwari
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Rakesh Trivedi
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shiaw-Yih Lin
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
198
|
Bouwman W, Verhaegh W, van de Stolpe A. Improved diagnosis of inflammatory bowel disease and prediction and monitoring of response to anti-TNF alpha treatment based on measurement of signal transduction pathway activity. Front Pharmacol 2022; 13:1008976. [PMID: 37090899 PMCID: PMC10115426 DOI: 10.3389/fphar.2022.1008976] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: Ulcerative colitis (UC) and Crohn’s disease (CD) are two subtypes of chronic inflammatory bowel disease (IBD). Differential diagnosis remains a challenge. Anti-TNFα treatment is an important treatment for IBD, yet resistance frequently occurs and cannot be predicted. Consequently, many patients receive ineffective therapy with potentially adverse effects. Novel assays are needed to improve diagnosis, and predict and monitor response to anti-TNF-α compounds.Design: Signal transduction pathway (STP) technology was used to quantify activity of STPs (androgen and estrogen receptor, PI3K, MAPK, TGFβ, Notch, Hedgehog, Wnt, NFκB, JAK-STAT1/2, and JAK-STAT3 pathways) in colon mucosa samples of CD and UC patients, based on transcriptome analysis. Previously described STP assay technology is based on computational inference of STP activity from mRNA levels of target genes of the STP transcription factor.Results: Results show that NFκB, JAK-STAT3, Wnt, MAPK, and androgen receptor pathways were abnormally active in CD and UC. Colon and ileum-localized CD differed with respect to STP activity, the JAK-STAT1/2 pathway being abnormally active in ileal CD. High activity of NFκB, JAK-STAT3, and TGFβ pathways was associated with resistance to anti-TNFα treatment in UC and colon-located CD, but not in ileal CD. Abnormal STP activity decreased with successful treatment.Conclusion: We believe that measuring mucosal STP activity provides clinically relevant information to improve differential diagnosis of IBD and prediction of resistance to anti-TNFα treatment in patients with colon-localized IBD, and provides new targets for treatment and overcoming anti-TNFα resistance.
Collapse
|
199
|
Galizia D, Minei S, Maldi E, Chilà G, Polidori A, Merlano MC. How Risk Factors Affect Head and Neck Squamous Cell Carcinoma (HNSCC) Tumor Immune Microenvironment (TIME): Their Influence on Immune Escape Mechanisms and Immunotherapy Strategy. Biomedicines 2022; 10:biomedicines10102498. [PMID: 36289760 PMCID: PMC9599463 DOI: 10.3390/biomedicines10102498] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/30/2022] [Accepted: 10/02/2022] [Indexed: 12/24/2022] Open
Abstract
Most head and neck squamous cell carcinomas (HNSCCs) are caused by lifestyle, such as cigarette smoking, or by viruses, such as human papillomavirus (HPV) and Epstein–Barr virus (EBV). HNSCC remains a clinical challenge, notwithstanding the improvements observed in the past years, involving surgery, radiotherapy, and chemotherapy. Recurrent/metastatic (R/M) disease represents an unmet clinical need. Immunotherapy has improved the prognosis of a small proportion of these patients, but most still do not benefit. In the last decade, several preclinical and clinical studies have explored the HNSCC tumor immune microenvironment (TIME), identifying important differences between smoking-associated and virus-associated HNSCCs. This review aims to present how different etiologies affect the HNSCC TIME, affecting immune escape mechanisms and sensitivity to immunotherapy.
Collapse
Affiliation(s)
- Danilo Galizia
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
- Correspondence:
| | - Silvia Minei
- Post-Graduate School of Specialization in Medical Oncology, University of Bari ‘A. Moro’, 70120 Bari, Italy
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, 70120 Bari, Italy
| | - Elena Maldi
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
| | - Giovanna Chilà
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
| | | | | |
Collapse
|
200
|
Wei T, Richter GT, Zhang H, Sun RW, Smith CH, Strub GM. Extracranial arteriovenous malformations demonstrate dysregulated TGF-β/BMP signaling and increased circulating TGF-β1. Sci Rep 2022; 12:16612. [PMID: 36198763 PMCID: PMC9534897 DOI: 10.1038/s41598-022-21217-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 09/23/2022] [Indexed: 12/03/2022] Open
Abstract
Extracranial arteriovenous malformations (AVMs) are characterized by anomalous arterial-to-venous connections, aberrant angiogenesis, local inflammation and hypoxia, and disorganized histological architecture; however, the precise molecular perturbations leading to this phenotype remain elusive. We hypothesized that extracranial AVM tissue would demonstrate deregulation of the TGF-β/BMP signaling pathway, which may serve as a potential target in the development of molecular-based therapies for AVMs. AVM tissue was harvested during resection from 10 patients with AVMs and compared to control tissue. Blood was collected from 14 AVM patients and 10 patients without AVMs as controls. Expression of TGF-β/BMP pathway components was analyzed using RT-PCR, western blotting, and immunohistochemistry. Circulating levels of TGF-β1 were analyzed by ELISA. Paired t tests were utilized to perform statistical analysis. The mRNA levels of TGF-β1, ALK1, Endoglin (ENG), Smad6, Smad7, and Smad8 were significantly elevated in AVM tissue when compared to controls. Protein levels of TGF-β1 and Smad3 were elevated in AVM tissue while protein levels of BMP-9, ALK1, Smad1, Smad6, and Smad8 were significantly decreased in AVMs. Immunohistochemistry demonstrated increased TGF-β1 in the perivascular cells of AVMs compared to normal controls, and circulating levels of TGF-β1 were significantly higher in AVM patients. Patients with AVMs demonstrate aberrant TGF-β/BMP expression in AVM tissue and blood compared to controls. Targeting aberrantly expressed components of the TGF-β/BMP pathway in extracranial AVMs may be a viable approach in the development of novel molecular therapies, and monitoring circulating TGF-β1 levels may be a useful indicator of treatment success.
Collapse
Affiliation(s)
- Ting Wei
- Arkansas Children's Research Institute, 13 Children's Way, Little Rock, AR, 72202, USA
| | - Gresham T Richter
- Arkansas Children's Research Institute, 13 Children's Way, Little Rock, AR, 72202, USA.,Department of Otolaryngology, University of Arkansas for Medical Sciences, 4301 W. Markham St., Little Rock, AR, 72205, USA
| | - Haihong Zhang
- Arkansas Children's Research Institute, 13 Children's Way, Little Rock, AR, 72202, USA.,Department of Otolaryngology, University of Arkansas for Medical Sciences, 4301 W. Markham St., Little Rock, AR, 72205, USA
| | - Ravi W Sun
- Arkansas Children's Research Institute, 13 Children's Way, Little Rock, AR, 72202, USA.,Department of Otolaryngology, University of Arkansas for Medical Sciences, 4301 W. Markham St., Little Rock, AR, 72205, USA
| | - Conor H Smith
- Department of Otolaryngology, University of Arkansas for Medical Sciences, 4301 W. Markham St., Little Rock, AR, 72205, USA
| | - Graham M Strub
- Arkansas Children's Research Institute, 13 Children's Way, Little Rock, AR, 72202, USA. .,Department of Otolaryngology, University of Arkansas for Medical Sciences, 4301 W. Markham St., Little Rock, AR, 72205, USA.
| |
Collapse
|