201
|
Alfaleh MA, Alsaab HO, Mahmoud AB, Alkayyal AA, Jones ML, Mahler SM, Hashem AM. Phage Display Derived Monoclonal Antibodies: From Bench to Bedside. Front Immunol 2020; 11:1986. [PMID: 32983137 PMCID: PMC7485114 DOI: 10.3389/fimmu.2020.01986] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Monoclonal antibodies (mAbs) have become one of the most important classes of biopharmaceutical products, and they continue to dominate the universe of biopharmaceutical markets in terms of approval and sales. They are the most profitable single product class, where they represent six of the top ten selling drugs. At the beginning of the 1990s, an in vitro antibody selection technology known as antibody phage display was developed by John McCafferty and Sir. Gregory Winter that enabled the discovery of human antibodies for diverse applications, particularly antibody-based drugs. They created combinatorial antibody libraries on filamentous phage to be utilized for generating antigen specific antibodies in a matter of weeks. Since then, more than 70 phage–derived antibodies entered clinical studies and 14 of them have been approved. These antibodies are indicated for cancer, and non-cancer medical conditions, such as inflammatory, optical, infectious, or immunological diseases. This review will illustrate the utility of phage display as a powerful platform for therapeutic antibodies discovery and describe in detail all the approved mAbs derived from phage display.
Collapse
Affiliation(s)
- Mohamed A Alfaleh
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia.,Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Ahmad Bakur Mahmoud
- College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Almohanad A Alkayyal
- Department of Medical Laboratory Technology, University of Tabuk, Tabuk, Saudi Arabia
| | - Martina L Jones
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia.,Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Stephen M Mahler
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia.,Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Anwar M Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
202
|
Abstract
R-spondin (RSPO) proteins amplify Wnt signaling and stimulate regeneration in a variety of tissues. To repair tissue in a tissue-specific manner, tissue-targeted RSPO mimetic molecules are desired. Here, we mutated RSPO (RSPO2 F105R/F109A) to eliminate LGR binding while preserving ZNRF3/RNF43 binding and targeted the mutated RSPO to a liver specific receptor, ASGR1. The resulting bi-specific molecule (αASGR1-RSPO2-RA) enhanced Wnt signaling effectively in vitro, and its activity was limited to ASGR1 expressing cells. Systemic administration of αASGR1-RSPO2-RA in mice specifically upregulated Wnt target genes and stimulated cell proliferation in liver but not intestine (which is more responsive to non-targeted RSPO2) in healthy mice, and improved liver function in diseased mice. These results not only suggest that a tissue-specific RSPO mimetic protein can stimulate regeneration in a cell-specific manner, but also provide a blueprint of how a tissue-specific molecule might be constructed for applications in a broader context.
Collapse
|
203
|
Diamond JR, Becerra C, Richards D, Mita A, Osborne C, O'Shaughnessy J, Zhang C, Henner R, Kapoun AM, Xu L, Stagg B, Uttamsingh S, Brachmann RK, Farooki A, Mita M. Phase Ib clinical trial of the anti-frizzled antibody vantictumab (OMP-18R5) plus paclitaxel in patients with locally advanced or metastatic HER2-negative breast cancer. Breast Cancer Res Treat 2020; 184:53-62. [PMID: 32803633 DOI: 10.1007/s10549-020-05817-w] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 07/17/2020] [Indexed: 11/29/2022]
Abstract
PURPOSE Vantictumab is a monoclonal antibody that binds to frizzled (FZD) receptors and inhibits canonical WNT signaling. This phase Ib dose escalation study enrolled patients with locally recurrent or metastatic HER2-negative breast cancer who were treated with weekly paclitaxel in combination with escalating doses of vantictumab. METHODS Patients were enrolled in dose escalation cohorts treated with weekly paclitaxel 90 mg/m2 on days 1, 8 and 15 in combination with vantictumab 3.5-14 mg/kg days 1 and 15 or 3-8 mg/kg day 1 of every 28-day cycle. Primary endpoints were safety, dose-limiting toxicities (DLTs). Secondary endpoints included pharmacokinetics, efficacy and an exploratory biomarker analysis. RESULTS Forty-eight female patients with a mean age of 54 were enrolled. The majority (66.6%) received prior chemotherapy for recurrent or metastatic disease; 45.8% were hormone receptor (HR)-positive, HER2-negative and 54.2% triple-negative. The most frequent adverse events related to any study treatment were nausea (54.2%), alopecia (52.1%), fatigue (47.9%), and peripheral neuropathy (43.8%). No DLTs occurred; however, 6 patients experienced fractures outside of the DLT window. The overall response rate was 31.3% and the clinical benefit rate was 68.8%. A 6-gene WNT pathway signature showed significant association with progression-free survival (PFS) and overall survival (OS) for the biomarker high versus biomarker low groups (PFS: p = 0.029 and OS: p = 0.00045, respectively). CONCLUSIONS The combination of vantictumab and weekly paclitaxel was generally well tolerated with promising efficacy; however, the incidence of fractures limits future clinical development of this particular WNT inhibitor in metastatic breast cancer. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov registration: NCT01973309.
Collapse
Affiliation(s)
- Jennifer R Diamond
- University of Colorado Anschutz Medical Campus, University of Colorado Cancer Center, 12801 E 17th Ave, Mailstop 8117, Aurora, CO, 80045, USA.
| | - Carlos Becerra
- Baylor Sammons Cancer Center, Texas Oncology, US Oncology, Dallas, TX, USA
| | | | - Alain Mita
- Samuel Oschin Comprehensive Cancer Institute, 8700 Beverly Blvd, SCCT Mezzanine MS 35, Los Angeles, CA, 90048, USA
| | - Cynthia Osborne
- Baylor Sammons Cancer Center, Texas Oncology, US Oncology, Dallas, TX, USA
| | | | - Chun Zhang
- OncoMed Pharmaceuticals, Redwood City, CA, USA
| | | | | | - Lu Xu
- OncoMed Pharmaceuticals, Redwood City, CA, USA
| | - Bob Stagg
- OncoMed Pharmaceuticals, Redwood City, CA, USA
| | | | | | - Azeez Farooki
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Monica Mita
- Samuel Oschin Comprehensive Cancer Institute, 8700 Beverly Blvd, SCCT Mezzanine MS 35, Los Angeles, CA, 90048, USA.
| |
Collapse
|
204
|
Taherkhani F, Hosseini KM, Zebardast S, Chegini KG, Gheibi N. Anti proliferative and apoptotic effects on pancreatic cancer cell lines indicate new roles for ANGPTL8 (Betatrophin). Genet Mol Biol 2020; 43:e20190196. [PMID: 32745158 PMCID: PMC7416753 DOI: 10.1590/1678-4685-gmb-2019-0196] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 05/31/2020] [Indexed: 11/25/2022] Open
Abstract
Despite considerable advances, the treatment of pancreatic cancer (PC) still
requires much effort. Unusual regulation of the Wnt and apoptotic signaling
pathways is widespread in cancer incidence. For instance, the
WIF1 (Wnt inhibitory factor 1) gene is down-regulated in
many cancers. The purpose of this study was to determine the effects of
recombinant Betatrophin, a recently discovered hormone, on MiaPaca-II and
Panc-1 pancreatic cell lines. Various concentrations of
Betatrophin were added to MiaPaca-II and Panc-1 pancreatic cell
lines during periods of 24 , 48, and 72 h. The MTT assay was applied to
investigate cell proliferation after treatment. The rate of apoptotic cells was
assessed using double-staining flow cytometry, and the expression levels of the
WIF1 gene and Bcl2 protein was observed by real-time PCR
and western blotting, respectively. The findings of this study suggest that
Betatrophin has an anti-proliferative effect on both MiaPaca-II and Panc-1 cell
lines, in line with the up-regulation of WIF1 as a tumor
suppressor gene. Moreover, the induction of apoptosis by ANGPTL8 occurred by the
down-regulation of Bcl2. Thus, Betatrophin can be proposed as a potential
therapeutic drug for treating pancreatic cancer.
Collapse
Affiliation(s)
| | | | - Sanaz Zebardast
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Koorosh Goodarzvand Chegini
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Nematollah Gheibi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
205
|
Dotan E, Cardin DB, Lenz HJ, Messersmith W, O'Neil B, Cohen SJ, Denlinger CS, Shahda S, Astsaturov I, Kapoun AM, Brachmann RK, Uttamsingh S, Stagg RJ, Weekes C. Phase Ib Study of Wnt Inhibitor Ipafricept with Gemcitabine and nab-paclitaxel in Patients with Previously Untreated Stage IV Pancreatic Cancer. Clin Cancer Res 2020; 26:5348-5357. [PMID: 32694153 DOI: 10.1158/1078-0432.ccr-20-0489] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/01/2020] [Accepted: 07/17/2020] [Indexed: 12/27/2022]
Abstract
PURPOSE The recombinant fusion protein ipafricept blocks Wnt signaling, and in combination with gemcitabine and nab-paclitaxel caused tumor regression in xenografts. This phase Ib study evaluated the combination of ipafricept with nab-paclitaxel + gemcitabine in patients with untreated metastatic pancreatic adenocarcinoma (mPDAC). PATIENTS AND METHODS Dose escalation started with standard dose nab-paclitaxel + gemcitabine and ipafricept (3.5 mg/kg days 1, 15). Because of fragility fractures seen with different anti-Wnt agents, following cohorts had ≥6 patients treated with ipafricept 3 to 5 mg/kg on day 1, and included bone marker monitoring and prophylactic bisphosphonates as indicated. On the basis of preclinical data, sequential dosing was evaluated in cohort 4 (ipafricept day 1 followed nab-paclitaxel + gemcitabine day 3). Objectives included safety, MTD, recommended phase II dose, pharmacokinetics, immunogenicity, pharmacodynamics, and efficacy. RESULTS A total of 26 patients were enrolled, five in cohort 1 and seven each in cohorts 2-4. ipafricept-related adverse events (AEs) of any grade included fatigue, nausea, vomiting, anorexia, and pyrexia. ipafricept-related AEs grade ≥3 included two events of aspartate aminotransferase elevation, and one each of nausea, rash, vomiting, and leucopenia. No dose-limiting toxicities or fragility fractures were observed. Nine patients (34.6%) had partial response, 12 (46.2%) stable disease as best response, with clinical benefit rate of 81%. Median progression-free survival was 5.9 m [95% confidence interval (CI), 3.4-18.4], median overall survival was 9.7 m (95% CI, 7.0-14). The study was terminated by the sponsor due to bone-related toxicity within this therapeutic program and concerns for commercial viability. One patient remains on therapy under compassionate use. CONCLUSIONS Ipafricept can be administered with nab-paclitaxel + gemcitabine with reasonable tolerance. Wnt pathway remains a therapeutic target of interest in mPDAC.
Collapse
Affiliation(s)
- Efrat Dotan
- Fox Chase Cancer Center, Philadelphia, Pennsylvania.
| | - Dana B Cardin
- Vanderbilt University, Medical Center, Nashville, Tennessee
| | | | | | | | - Steven J Cohen
- Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | | | | | | | - Ann M Kapoun
- OncoMed Pharmaceuticals, Redwood City, California
| | | | | | | | - Colin Weekes
- Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
206
|
Gajos-Michniewicz A, Czyz M. WNT Signaling in Melanoma. Int J Mol Sci 2020; 21:E4852. [PMID: 32659938 PMCID: PMC7402324 DOI: 10.3390/ijms21144852] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
WNT-signaling controls important cellular processes throughout embryonic development and adult life, so any deregulation of this signaling can result in a wide range of pathologies, including cancer. WNT-signaling is classified into two categories: β-catenin-dependent signaling (canonical pathway) and β-catenin-independent signaling (non-canonical pathway), the latter can be further divided into WNT/planar cell polarity (PCP) and calcium pathways. WNT ligands are considered as unique directional growth factors that contribute to both cell proliferation and polarity. Origin of cancer can be diverse and therefore tissue-specific differences can be found in WNT-signaling between cancers, including specific mutations contributing to cancer development. This review focuses on the role of the WNT-signaling pathway in melanoma. The current view on the role of WNT-signaling in cancer immunity as well as a short summary of WNT pathway-related drugs under investigation are also provided.
Collapse
Affiliation(s)
| | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92–215 Lodz, Poland;
| |
Collapse
|
207
|
Chen S, Yuan X, Xu H, Yi M, Liu S, Wen F. WNT974 Inhibits Proliferation, Induces Apoptosis, and Enhances Chemosensitivity to Doxorubicin in Lymphoma Cells by Inhibiting Wnt/β-Catenin Signaling. Med Sci Monit 2020; 26:e923799. [PMID: 32597418 PMCID: PMC7346748 DOI: 10.12659/msm.923799] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Upregulation of the Wnt/β-catenin pathway has been demonstrated to promote tumor proliferation and chemoresistance in lymphoma. Our objective was to evaluate the effect of the Wnt/β-catenin pathway inhibitor WNT974 in lymphoma cells. Material/Methods Human lymphoma cell lines HUT-78 and BJAB were treated with or without 1 μM WNT974±0.15 μg/L doxorubicin (Dox). Cell viability and proliferation were evaluated by CCK-8 and colony formation assay. Expression of proliferating cell nuclear antigen (PCNA), KI67, and apoptotic-related proteins including Bcl-2, Bax, cleaved-caspase3, and cleaved-caspase9, together with Wnt pathway proteins Wnt, β-catenin, Axin2, and c-Myc, were detected by Western blot analysis. Flow cytometry was used to calculate the ratio of apoptotic cells. Results In HUT-78 and BJAB cells, 1 μM WNT974 significantly reduced viability and colony formation. The expression of 2 markers of tumor cell proliferation, protein PCNA and KI67, was also reduced by WNT974. Treatment with 1 μM WNT974 for 48 h increased the rate of cell apoptosis, inhibited the expression of anti-apoptotic protein Bcl-2, and enhanced pro-apoptotic proteins Bax, cleaved-caspase3, and cleaved-caspase9 expression in both cell lines. After treatment with WNT974 plus Dox, cell viability was markedly decreased compared with Dox treatment alone. Mechanistically, WNT974 prevented the expression of Wnt, Axin2, β-catenin, and its target gene c-Myc. Conclusions WNT974 effectively treats lymphoma by inhibiting cell proliferation, inducing cell apoptosis, and enhancing chemosensitivity to Dox, and these effects are dependent on blocking Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Senmin Chen
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, Guangdong, China (mainland)
| | - Xiuli Yuan
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, Guangdong, China (mainland)
| | - Huanli Xu
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, Guangdong, China (mainland)
| | - Meng Yi
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, Guangdong, China (mainland)
| | - Sixi Liu
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, Guangdong, China (mainland)
| | - Feiqiu Wen
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, Guangdong, China (mainland)
| |
Collapse
|
208
|
Aghabozorgi AS, Ebrahimi R, Bahiraee A, Tehrani SS, Nabizadeh F, Setayesh L, Jafarzadeh-Esfehani R, Ferns GA, Avan A, Rashidi Z. The genetic factors associated with Wnt signaling pathway in colorectal cancer. Life Sci 2020; 256:118006. [PMID: 32593708 DOI: 10.1016/j.lfs.2020.118006] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022]
Abstract
Colorectal cancer (CRC) is a common cancer with poor prognosis and high mortality. There is growing information about the factors involved in the pathogenesis of CRC. However, the knowledge of the predisposing factors is limited. The development of CRC is strongly associated with the Wingless/Integrated (Wnt) signaling pathway. This pathway comprises several major target proteins, including LRP5/6, GSK3β, adenomatous polyposis coli (APC), axis inhibition protein (Axin), and β-catenin. Genetic variations in these components of the Wnt signaling pathway may lead to the activation of β-catenin, potentially increasing the proliferation of colorectal cells. Because of the potentially important role of the Wnt signaling pathway in CRC, we aimed to review the involvement of different mutations in the main downstream proteins of this pathway, including LRP5/6, APC, GSK3β, Axin, and β-catenin. Determination of the genetic risk factors involved in the progression of CRC may lead to novel approaches for the early diagnosis of CRC and the identification of potential therapeutic targets in the treatment of CRC.
Collapse
Affiliation(s)
- Amirsaeed Sabeti Aghabozorgi
- Medical Genetics Research Center, Basic Medical Sciences Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reyhane Ebrahimi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Bahiraee
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Sadra Samavarchi Tehrani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Nabizadeh
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Leila Setayesh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran; Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Jafarzadeh-Esfehani
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Amir Avan
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Zahra Rashidi
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Anatomical Sciences, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
209
|
Wnt Signaling in Gynecologic Malignancies. Int J Mol Sci 2020; 21:ijms21124272. [PMID: 32560059 PMCID: PMC7348953 DOI: 10.3390/ijms21124272] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/12/2020] [Accepted: 06/13/2020] [Indexed: 12/24/2022] Open
Abstract
Gynecologic malignancies, including ovarian cancer, endometrial cancer, and cervical cancer, affect hundreds of thousands of women worldwide every year. Wnt signaling, specifically Wnt/β-catenin signaling, has been found to play an essential role in many oncogenic processes in gynecologic malignancies, including tumorigenesis, metastasis, recurrence, and chemotherapy resistance. As such, the Wnt/β-catenin signaling pathway has the potential to be a target for effective treatment, improving patient outcomes. In this review, we discuss the evidence supporting the importance of the Wnt signaling pathways in the development, progression, and treatment of gynecologic malignancies.
Collapse
|
210
|
Rim EY, Kinney LK, Nusse R. β-catenin-mediated Wnt signal transduction proceeds through an endocytosis-independent mechanism. Mol Biol Cell 2020; 31:1425-1436. [PMID: 32320321 PMCID: PMC7353137 DOI: 10.1091/mbc.e20-02-0114] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/07/2020] [Accepted: 04/14/2020] [Indexed: 01/12/2023] Open
Abstract
The Wnt pathway is a key intercellular signaling cascade that regulates development, tissue homeostasis, and regeneration. However, gaps remain in our understanding of the molecular events that take place between ligand-receptor binding and target gene transcription. We used a novel tool for quantitative, real-time assessment of endogenous pathway activation, measured in single cells, to answer an unresolved question in the field-whether receptor endocytosis is required for Wnt signal transduction. We combined knockdown or knockout of essential components of clathrin-mediated endocytosis with quantitative assessment of Wnt signal transduction in mouse embryonic stem cells (mESCs). Disruption of clathrin-mediated endocytosis did not affect accumulation and nuclear translocation of β-catenin, as measured by single-cell live imaging of endogenous β-catenin, and subsequent target gene transcription. Disruption of another receptor endocytosis pathway, caveolin-mediated endocytosis, did not affect Wnt pathway activation in mESCs. Additional results in multiple cell lines support that endocytosis is not a requirement for Wnt signal transduction. We show that off-target effects of a drug used to inhibit endocytosis may be one source of the discrepancy among reports on the role of endocytosis in Wnt signaling.
Collapse
Affiliation(s)
- Ellen Youngsoo Rim
- Howard Hughes Medical Institute, Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Leigh Katherine Kinney
- Howard Hughes Medical Institute, Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Roeland Nusse
- Howard Hughes Medical Institute, Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
211
|
Jiang Y, Zhuo X, Mao C. G Protein-coupled Receptors in Cancer Stem Cells. Curr Pharm Des 2020; 26:1952-1963. [DOI: 10.2174/1381612826666200305130009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/10/2020] [Indexed: 12/19/2022]
Abstract
G protein-coupled receptors (GPCRs) are highly expressed on a variety of tumour tissues while several
GPCR exogenous ligands become marketed pharmaceuticals. In recent decades, cancer stem cells (CSCs) become
widely investigated drug targets for cancer therapy but the underlying mechanism is still not fully elucidated.
There are vigorous participations of GPCRs in CSCs-related signalling and functions, such as biomarkers for
CSCs, activation of Wnt, Hedgehog (HH) and other signalling to facilitate CSCs progressions. This relationship
can not only uncover a novel molecular mechanism for GPCR-mediated cancer cell functions but also assist our
understanding of maintaining and modulating CSCs. Moreover, GPCR antagonists and monoclonal antibodies
could be applied to impair CSCs functions and consequently attenuate tumour growth, some of which have been
undergoing clinical studies and are anticipated to turn into marketed anticancer drugs. Therefore, this review
summarizes and provides sufficient evidences on the regulation of GPCR signalling in the maintenance, differentiation
and pluripotency of CSCs, suggesting that targeting GPCRs on the surface of CSCs could be potential
therapeutic strategies for cancer therapy.
Collapse
Affiliation(s)
- Yuhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Xin Zhuo
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Canquan Mao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| |
Collapse
|
212
|
Davis SL, Cardin DB, Shahda S, Lenz HJ, Dotan E, O'Neil BH, Kapoun AM, Stagg RJ, Berlin J, Messersmith WA, Cohen SJ. A phase 1b dose escalation study of Wnt pathway inhibitor vantictumab in combination with nab-paclitaxel and gemcitabine in patients with previously untreated metastatic pancreatic cancer. Invest New Drugs 2020; 38:821-830. [PMID: 31338636 PMCID: PMC7211194 DOI: 10.1007/s10637-019-00824-1] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 06/19/2019] [Indexed: 01/05/2023]
Abstract
Vantictumab is a fully human monoclonal antibody that inhibits Wnt pathway signaling through binding FZD1, 2, 5, 7, and 8 receptors. This phase Ib study evaluated vantictumab in combination with nab-paclitaxel and gemcitabine in patients with untreated metastatic pancreatic adenocarcinoma. Patients received vantictumab at escalating doses in combination with standard dosing of nab-paclitaxel and gemcitabine according to a 3 + 3 design. A total of 31 patients were treated in 5 dosing cohorts. Fragility fractures attributed to vantictumab occurred in 2 patients in Cohort 2 (7 mg/kg every 2 weeks), and this maximum administered dose (MAD) on study was considered unsafe. The dosing schedule was revised to every 4 weeks for Cohorts 3 through 5, with additional bone safety parameters added. Sequential dosing of vantictumab followed by nab-paclitaxel and gemcitabine was also explored. No fragility fractures attributed to vantictumab occurred in these cohorts; pathologic fracture not attributed to vantictumab was documented in 2 patients. The study was ultimately terminated due to concerns around bone-related safety, and thus the maximum tolerated dose (MTD) of the combination was not determined. The MAD of vantictumab according to the revised dosing schedule was 5 mg/kg (n = 16).
Collapse
Affiliation(s)
| | | | | | | | - Efrat Dotan
- Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | | | | | | | | | - Steven J Cohen
- Jefferson Health/Abington Memorial Hospital, Abington, PA, USA
| |
Collapse
|
213
|
Manandhar S, Kabekkodu SP, Pai KSR. Aberrant canonical Wnt signaling: Phytochemical based modulation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 76:153243. [PMID: 32535482 DOI: 10.1016/j.phymed.2020.153243] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 04/07/2020] [Accepted: 05/10/2020] [Indexed: 05/26/2023]
Abstract
BACKGROUND Wnt signaling pathway plays a major role during development like gastrulation, axis formation, organ development and organization of body plan development. Wnt signaling aberration has been linked with various disease conditions like osteoporosis, colon cancer, hair follicle tumor, Leukemia, and Alzheimer's disease. Phytochemicals like flavonoid, glycosides, polyphenols, have been reported to directly target the markers of Wnt signaling in different disease models. PURPOSE The study deals in detail about the different phytochemical targeting key players of Wnt signaling pathway in diseases like Cancer, Osteoporosis, and Alzheimer's disease. We have focused on the Pharmacological basis of disease alleviation by phytochemical specifically targeting the Wnt signaling markers in this study. METHODS The study focused on the published articles from the preclinical rodent and invitro cell line studies related to Wnt signaling and Phytochemicals related to Cancer, Alzheimer's and Osteoporosis. The electronic databases Scopus, Web of Science and Pubmed database were used for the systematic search of literatures from 2005 up to 2019 using keywords Canonical Wnt signaling pathway, Cancer, Alzheimer's disease, Osteoporosis, Phytochemicals. The focus was to identify the target specific modulation of Wnt signaling mediated by phytochemicals. RESULTS Approximately 30 phytochemicals of different class have been identified to modulate Wnt signaling pathway acting through Axin, β-catenin translocation, GSK-3β, AKT, Wif-1 in various experimental studies. The down regulation of Wnt signaling is observed in Cancer mostly colorectal cancer, breast cancer mediated through mutations in APC and Axin genes. Different class of Phytochemicals such as flavonoid, glycosides, polyphenol, alkaloids etc. have been found to target Wnt signaling markers and alleviate Cancer. Similarly, Up regulation of Wnt signaling has been reported in Osteoporosis and neurodegenerative disease like Alzheimer's disease. CONCLUSION This review highlights the possibility of the Phytochemicals to target Wnt markers and its potential to either activate or deactivate the Wnt signaling pathway. It also describes the challenges in proper targeting of Wnt signaling and the potential risk and consequences of either up regulation or down regulation of the signaling pathway. This article highlights the possibility of Wnt signaling pathway as a therapeutic option in different diseases.
Collapse
Affiliation(s)
- Suman Manandhar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - K Sreedhara Ranganath Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India.
| |
Collapse
|
214
|
Shuck SC, Nguyen C, Chan Y, O’Connor T, Ciminera AK, Kahn M, Termini J. Metal-Assisted Protein Quantitation (MAPq): Multiplex Analysis of Protein Expression Using Lanthanide-Modified Antibodies with Detection by Inductively Coupled Plasma Mass Spectrometry. Anal Chem 2020; 92:7556-7564. [DOI: 10.1021/acs.analchem.0c00058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
215
|
Li X, Larsson P, Ljuslinder I, Öhlund D, Myte R, Löfgren-Burström A, Zingmark C, Ling A, Edin S, Palmqvist R. Ex Vivo Organoid Cultures Reveal the Importance of the Tumor Microenvironment for Maintenance of Colorectal Cancer Stem Cells. Cancers (Basel) 2020; 12:E923. [PMID: 32290033 PMCID: PMC7226030 DOI: 10.3390/cancers12040923] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022] Open
Abstract
Colorectal cancer (CRC) is a heterogeneous disease, with varying clinical presentations and patient prognosis. Different molecular subgroups of CRC should be treated differently and therefore, must be better characterized. Organoid culture has recently been suggested as a good model to reflect the heterogeneous nature of CRC. However, organoid cultures cannot be established from all CRC tumors. The study examines which CRC tumors are more likely to generate organoids and thus benefit from ex vivo organoid drug testing. Long-term organoid cultures from 22 out of 40 CRC tumor specimens were established. It was found that organoid cultures were more difficult to establish from tumors characterized as microsatellite instable (MSI), BRAF-mutated, poorly differentiated and/or of a mucinous type. This suggests that patients with such tumors are less likely to benefit from ex vivo organoid drug testing, but it may also suggest biological difference in tumor growth. RNA sequencing analysis of tumor sections revealed that the in vivo maintenance of these non-organoid-forming tumors depends on factors related to inflammation and pathogen exposure. Furthermore, using TCGA data we could show a trend towards a worse prognosis for patients with organoid-forming tumors, suggesting also clinical differences. Results suggest that organoids are more difficult to establish from tumors characterized as MSI, BRAF-mutated, poorly differentiated and/or of a mucinous type. We further suggest that the maintenance of cell growth of these tumors in vivo may be promoted by immune-related factors and other stromal components within the tumor microenvironment.
Collapse
Affiliation(s)
- Xingru Li
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| | - Pär Larsson
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| | - Ingrid Ljuslinder
- Department of Radiation Sciences, Oncology, Umeå University, 90185 Umeå, Sweden; (I.L.); (D.O.); (R.M.)
| | - Daniel Öhlund
- Department of Radiation Sciences, Oncology, Umeå University, 90185 Umeå, Sweden; (I.L.); (D.O.); (R.M.)
- Wallenberg Center for Molecular Medicine, Umeå University, 90185 Umeå, Sweden
| | - Robin Myte
- Department of Radiation Sciences, Oncology, Umeå University, 90185 Umeå, Sweden; (I.L.); (D.O.); (R.M.)
| | - Anna Löfgren-Burström
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| | - Carl Zingmark
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| | - Agnes Ling
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| | - Sofia Edin
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| | - Richard Palmqvist
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| |
Collapse
|
216
|
Chen H, Lu C, Ouyang B, Zhang H, Huang Z, Bhatia D, Lee SJ, Shah D, Sura A, Yeh WC, Li Y. Development of Potent, Selective Surrogate WNT Molecules and Their Application in Defining Frizzled Requirements. Cell Chem Biol 2020; 27:598-609.e4. [PMID: 32220333 DOI: 10.1016/j.chembiol.2020.02.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 02/10/2020] [Accepted: 02/27/2020] [Indexed: 01/20/2023]
Abstract
WNTs regulate myriad biological processes during embryonic development and are key regulators of stem cell function, tissue homeostasis, and injury repair in adults. The creation of WNT-based therapies has been hampered by challenges in developing soluble, potent, and selective WNT molecules. Soluble WNT surrogates have been reported, but they demonstrate relatively weak WNT signaling activity. Here, we describe a platform for potent, selective WNT surrogate generation. We identify multivalent binding to Frizzleds (FZDs) and low-density lipoprotein receptor-related proteins (LRPs) to be a requirement for maximal WNT/β-catenin activation. Furthermore, we show that recruitment of two different FZDs together with LRP causes efficient signaling. Surrogate WNT targeting either FZD1,2,7 or FZD5,8 induces expansive growth of intestinal organoids. This flexible WNT surrogate platform yields potent agonists with any desired receptor specificity and will be useful for research and therapeutic applications for tissue regeneration.
Collapse
Affiliation(s)
- Hui Chen
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Chenggang Lu
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Brian Ouyang
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Haili Zhang
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Zhong Huang
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Diksha Bhatia
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Sung-Jin Lee
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Darshini Shah
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Asmiti Sura
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Wen-Chen Yeh
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Yang Li
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA.
| |
Collapse
|
217
|
Zhao Y, Ren J, Hillier J, Lu W, Jones EY. Antiepileptic Drug Carbamazepine Binds to a Novel Pocket on the Wnt Receptor Frizzled-8. J Med Chem 2020; 63:3252-3260. [PMID: 32049522 PMCID: PMC7104226 DOI: 10.1021/acs.jmedchem.9b02020] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Indexed: 01/07/2023]
Abstract
Misregulation of Wnt signaling is common in human cancer. The development of small molecule inhibitors against the Wnt receptor, frizzled (FZD), may have potential in cancer therapy. During small molecule screens, we observed binding of carbamazepine to the cysteine-rich domain (CRD) of the Wnt receptor FZD8 using surface plasmon resonance (SPR). Cellular functional assays demonstrated that carbamazepine can suppress FZD8-mediated Wnt/β-catenin signaling. We determined the crystal structure of the complex at 1.7 Å resolution, which reveals that carbamazepine binds at a novel pocket on the FZD8 CRD. The unique residue Tyr52 discriminates FZD8 from the closely related FZD5 and other FZDs for carbamazepine binding. The first small molecule-bound FZD structure provides a basis for anti-FZD drug development. Furthermore, the observed carbamazepine-mediated Wnt signaling inhibition may help to explain the phenomenon of bone loss and increased adipogenesis in some patients during long-term carbamazepine treatment.
Collapse
Affiliation(s)
- Yuguang Zhao
- Division of Structural
Biology,
Wellcome Centre for Human Genetics, University
of Oxford, Oxford OX3 7BN, United Kingdom
| | - Jingshan Ren
- Division of Structural
Biology,
Wellcome Centre for Human Genetics, University
of Oxford, Oxford OX3 7BN, United Kingdom
| | - James Hillier
- Division of Structural
Biology,
Wellcome Centre for Human Genetics, University
of Oxford, Oxford OX3 7BN, United Kingdom
| | - Weixian Lu
- Division of Structural
Biology,
Wellcome Centre for Human Genetics, University
of Oxford, Oxford OX3 7BN, United Kingdom
| | - E. Yvonne Jones
- Division of Structural
Biology,
Wellcome Centre for Human Genetics, University
of Oxford, Oxford OX3 7BN, United Kingdom
| |
Collapse
|
218
|
Otero-Ramirez ME, Matoba K, Mihara E, Passioura T, Takagi J, Suga H. Macrocyclic peptides that inhibit Wnt signalling via interaction with Wnt3a. RSC Chem Biol 2020; 1:26-34. [PMID: 34458746 PMCID: PMC8382136 DOI: 10.1039/d0cb00016g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 03/13/2020] [Indexed: 12/20/2022] Open
Abstract
Here we report de novo macrocyclic peptide binders to Wnt3a, a member of the Wnt protein family. By means of the Random non-standard Peptides Integrated Discovery (RaPID) system, we have performed in vitro selection against the complex of mouse Wnt3a (mWnt3a) with human afamin (hAFM) to discover macrocyclic peptides that bind mWnt3a with K D values as tight as 110 nM. One of these peptides, WAp-D04 (Wnt-AFM-peptide-D04), was able to inhibit the receptor-mediated signaling process, which was demonstrated in a Wnt3a-dependent reporter cell-line. Based on this initial hit, we applied a block-mutagenesis scanning display to identify a mutant inhibitor, WAp-D04-W10P, with 5-fold greater potency in a reporter assay. This work represents the first instance of molecules capable of inhibiting Wnt signaling through direct interaction with a Wnt protein, a molecular class for which targeting has been challenging due its highly hydrophobic nature.
Collapse
Affiliation(s)
- Manuel E Otero-Ramirez
- Department of Chemistry, Graduate School of Science, The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
| | - Kyoko Matoba
- Laboratory of Protein Synthesis and Expression, Institute for Protein Research, Osaka University 3-2 Yamadaoka Suita-shi Osaka 565-0871 Japan
| | - Emiko Mihara
- Laboratory of Protein Synthesis and Expression, Institute for Protein Research, Osaka University 3-2 Yamadaoka Suita-shi Osaka 565-0871 Japan
| | - Toby Passioura
- Department of Chemistry, Graduate School of Science, The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan .,Sydney Analytical, School of Chemistry and School of Life and Environmental Sciences, The University of Sydney Sydney 2006 Australia
| | - Junichi Takagi
- Laboratory of Protein Synthesis and Expression, Institute for Protein Research, Osaka University 3-2 Yamadaoka Suita-shi Osaka 565-0871 Japan
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
| |
Collapse
|
219
|
Triple-Negative Breast Cancer: A Review of Conventional and Advanced Therapeutic Strategies. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17062078. [PMID: 32245065 PMCID: PMC7143295 DOI: 10.3390/ijerph17062078] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 12/14/2022]
Abstract
Triple-negative breast cancer (TNBC) cells are deficient in estrogen, progesterone and ERBB2 receptor expression, presenting a particularly challenging therapeutic target due to their highly invasive nature and relatively low response to therapeutics. There is an absence of specific treatment strategies for this tumor subgroup, and hence TNBC is managed with conventional therapeutics, often leading to systemic relapse. In terms of histology and transcription profile these cancers have similarities to BRCA-1-linked breast cancers, and it is hypothesized that BRCA1 pathway is non-functional in this type of breast cancer. In this review article, we discuss the different receptors expressed by TNBC as well as the diversity of different signaling pathways targeted by TNBC therapeutics, for example, Notch, Hedgehog, Wnt/b-Catenin as well as TGF-beta signaling pathways. Additionally, many epidermal growth factor receptor (EGFR), poly (ADP-ribose) polymerase (PARP) and mammalian target of rapamycin (mTOR) inhibitors effectively inhibit the TNBCs, but they face challenges of either resistance to drugs or relapse. The resistance of TNBC to conventional therapeutic agents has helped in the advancement of advanced TNBC therapeutic approaches including hyperthermia, photodynamic therapy, as well as nanomedicine-based targeted therapeutics of drugs, miRNA, siRNA, and aptamers, which will also be discussed. Artificial intelligence is another tool that is presented to enhance the diagnosis of TNBC.
Collapse
|
220
|
Li X, Ortiz MA, Kotula L. The physiological role of Wnt pathway in normal development and cancer. Exp Biol Med (Maywood) 2020; 245:411-426. [PMID: 31996036 PMCID: PMC7082880 DOI: 10.1177/1535370220901683] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Over the decades, many studies have illustrated the critical roles of Wnt signaling pathways in both developmental processes as well as tumorigenesis. Due to the complexity of Wnt signaling regulation, there are still questions to be addressed about ways cells are able to manipulate different types of Wnt pathways in order to fulfill the requirements for normal or cancer development. In this review, we will describe different types of Wnt signaling pathways and their roles in both normal developmental processes and their role in cancer development and progression. Additionally, we will briefly introduce new strategies currently in clinical trials targeting Wnt signaling pathway components for cancer therapy.
Collapse
Affiliation(s)
- Xiang Li
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Maria A Ortiz
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Leszek Kotula
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
221
|
Zhong Z, Virshup DM. Wnt Signaling and Drug Resistance in Cancer. Mol Pharmacol 2020; 97:72-89. [PMID: 31787618 DOI: 10.1124/mol.119.117978] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/21/2019] [Indexed: 12/22/2022] Open
Abstract
Wnts are secreted proteins that bind to cell surface receptors to activate downstream signaling cascades. Normal Wnt signaling plays key roles in embryonic development and adult tissue homeostasis. The secretion of Wnt ligands, the turnover of Wnt receptors, and the signaling transduction are tightly regulated and fine-tuned to keep the signaling output "just right." Hyperactivated Wnt signaling due to recurrent genetic alterations drives several human cancers. Elevated Wnt signaling also confers resistance to multiple conventional and targeted cancer therapies through diverse mechanisms including maintaining the cancer stem cell population, enhancing DNA damage repair, facilitating transcriptional plasticity, and promoting immune evasion. Different classes of Wnt signaling inhibitors targeting key nodes of the pathway have been developed and show efficacy in treating Wnt-driven cancers and subverting Wnt-mediated therapy resistance in preclinical studies. Several of these inhibitors have advanced to clinical trials, both singly and in combination with other existing US Food and Drug Administration-approved anti-cancer modalities. In the near future, pharmacological inhibition of Wnt signaling may be a real choice for patients with cancer. SIGNIFICANCE STATEMENT: The latest insights in Wnt signaling, ranging from basic biology to therapeutic implications in cancer, are reviewed. Recent studies extend understanding of this ancient signaling pathway and describe the development and improvement of anti-Wnt therapeutic modalities for cancer.
Collapse
Affiliation(s)
- Zheng Zhong
- Department of Physiology, National University of Singapore, Singapore, Singapore (Z.Z.); Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore (Z.Z., D.M.V.); and Department of Pediatrics, Duke University, Durham, North Carolina (D.M.V.)
| | - David M Virshup
- Department of Physiology, National University of Singapore, Singapore, Singapore (Z.Z.); Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore (Z.Z., D.M.V.); and Department of Pediatrics, Duke University, Durham, North Carolina (D.M.V.)
| |
Collapse
|
222
|
van Schie EH, van Amerongen R. Aberrant WNT/CTNNB1 Signaling as a Therapeutic Target in Human Breast Cancer: Weighing the Evidence. Front Cell Dev Biol 2020; 8:25. [PMID: 32083079 PMCID: PMC7005411 DOI: 10.3389/fcell.2020.00025] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/14/2020] [Indexed: 12/22/2022] Open
Abstract
WNT signaling is crucial for tissue morphogenesis during development in all multicellular animals. After birth, WNT/CTNNB1 responsive stem cells are responsible for tissue homeostasis in various organs and hyperactive WNT/CTNNB1 signaling is observed in many different human cancers. The first link between WNT signaling and breast cancer was established almost 40 years ago, when Wnt1 was identified as a proto-oncogene capable of driving mammary tumor formation in mice. Since that discovery, there has been a dedicated search for aberrant WNT signaling in human breast cancer. However, much debate and controversy persist regarding the importance of WNT signaling for the initiation, progression or maintenance of different breast cancer subtypes. As the first drugs designed to block functional WNT signaling have entered clinical trials, many questions about the role of aberrant WNT signaling in human breast cancer remain. Here, we discuss three major research gaps in this area. First, we still lack a basic understanding of the function of WNT signaling in normal human breast development and physiology. Second, the overall extent and precise effect of (epi)genetic changes affecting the WNT pathway in different breast cancer subtypes are still unknown. Which underlying molecular and cell biological mechanisms are disrupted as a result also awaits further scrutiny. Third, we survey the current status of targeted therapeutics that are aimed at interfering with the WNT pathway in breast cancer patients and highlight the importance and complexity of selecting the subset of patients that may benefit from treatment.
Collapse
Affiliation(s)
| | - Renée van Amerongen
- Section of Molecular Cytology and van Leeuwenhoek Centre for Advanced Microscopy, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
223
|
Jiang N, Zou C, Zhu Y, Luo Y, Chen L, Lei Y, Tang K, Sun Y, Zhang W, Li S, He Q, Zhou J, Chen Y, Luo J, Jiang W, Ke Z. HIF-1ɑ-regulated miR-1275 maintains stem cell-like phenotypes and promotes the progression of LUAD by simultaneously activating Wnt/β-catenin and Notch signaling. Am J Cancer Res 2020; 10:2553-2570. [PMID: 32194819 PMCID: PMC7052895 DOI: 10.7150/thno.41120] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 01/05/2020] [Indexed: 02/07/2023] Open
Abstract
Rationale: Cancer stem cells (CSCs) are considered to be essential for tumorigenesis, recurrence, and metastasis and therefore serve as a biomarker for tumor progression in diverse cancers. Recent studies have illustrated that specific miRNAs exhibit novel therapeutic potential by controlling CSC properties. miR-1275 is upregulated in lung adenocarcinoma (LUAD) and enhances its stemness. However, the underlying mechanisms have not been elucidated. Methods: miRNA expression microarray of LUAD and adjacent nontumor tissues was used to identify miRNAs involved in LUAD malignant progression. miR-1275 expression level was determined using quantitative real-time PCR (RT-qPCR) and in situ hybridization (ISH), and its correlation with clinicopathological characteristics was analyzed in LUAD specimens. The upstream regulator of miR-1275 was validated by chromatin immunoprecipitation (ChIP). The biological functions and underlying mechanisms of miR-1275 were investigated both in vitro and in vivo. Results: MiR-1275 was highly upregulated in lung cancer cell lines and LUAD tissues. Overexpression of miR-1275 in lung cancer patients was associated with shorter overall- and recurrence-free-survival. Proto-oncogene HIF-1ɑ was identified as the transcription mediator of miR-1275. Activation of Wnt/β-catenin and Notch signaling by miR-1275 was found to enhance the stemness of LUAD cells, while antagonizing miR-1275 or suppressing Wnt/β-catenin and Notch pathways potently reversed miR-1275-induced pathway co-activation and stemness. Enhanced stemness dramatically promoted tumorigenicity, recurrence, and metastasis. miR-1275 directly targeted multiple antagonists of Wnt/β-catenin and Notch pathways, including DKK3, SFRP1, GSK3β, RUNX3, and NUMB, respectively, which resulted in signaling activation. Conclusions: Our findings identified miR-1275 as a potential oncogene in LUAD that exerts its tumorigenic effect through co-activating Wnt/β-catenin and Notch signaling pathways. Thus, HIF-1ɑ-regulated miR-1275 might be a potential therapeutic target for LUAD.
Collapse
|
224
|
Chang LS, Kim M, Glinka A, Reinhard C, Niehrs C. The tumor suppressor PTPRK promotes ZNRF3 internalization and is required for Wnt inhibition in the Spemann organizer. eLife 2020; 9:51248. [PMID: 31934854 PMCID: PMC6996932 DOI: 10.7554/elife.51248] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 01/12/2020] [Indexed: 12/12/2022] Open
Abstract
A hallmark of Spemann organizer function is its expression of Wnt antagonists that regulate axial embryonic patterning. Here we identify the tumor suppressor Protein tyrosine phosphatase receptor-type kappa (PTPRK), as a Wnt inhibitor in human cancer cells and in the Spemann organizer of Xenopus embryos. We show that PTPRK acts via the transmembrane E3 ubiquitin ligase ZNRF3, a negative regulator of Wnt signaling promoting Wnt receptor degradation, which is also expressed in the organizer. Deficiency of Xenopus Ptprk increases Wnt signaling, leading to reduced expression of Spemann organizer effector genes and inducing head and axial defects. We identify a '4Y' endocytic signal in ZNRF3, which PTPRK maintains unphosphorylated to promote Wnt receptor depletion. Our discovery of PTPRK as a negative regulator of Wnt receptor turnover provides a rationale for its tumor suppressive function and reveals that in PTPRK-RSPO3 recurrent cancer fusions both fusion partners, in fact, encode ZNRF3 regulators. How human and other animals form distinct head- and tail-ends as embryos is a fundamental question in biology. The fertilized eggs of the African clawed frog (also known as Xenopus) become embryos and grow into tadpoles within two days. This rapid growth makes Xenopus particularly suitable as a model to study how animals with backbones form their body plans. In Xenopus embryos, a small group of cells known as the Spemann organizer plays a pivotal role in forming the body plan. It produces several enzymes known as Wnt inhibitors that repress a signal pathway known as Wnt signaling to determine the head- and tail-ends of the embryo. Chang, Kim et al. searched for new Wnt inhibitors in the Spemann organizer of Xenopus embryos. The experiments revealed that the Spemann organizer produced an enzyme known as PTPRK that was essential to permit the head-to-tail patterning of the brain. PTPRK inhibited Wnt signaling by activating another enzyme known as ZNRF3. Previous studies have shown that defects in Wnt signaling and in the activities of PTPRK and ZNRF3 are involved in colon cancer in mammals. Thus, these findings may help to develop new approaches for treating cancer in the future.
Collapse
Affiliation(s)
- Ling-Shih Chang
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Minseong Kim
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Andrey Glinka
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Carmen Reinhard
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany.,Institute of Molecular Biology (IMB), Mainz, Germany
| |
Collapse
|
225
|
Alfaleh MA, Alsaab HO, Mahmoud AB, Alkayyal AA, Jones ML, Mahler SM, Hashem AM. Phage Display Derived Monoclonal Antibodies: From Bench to Bedside. Front Immunol 2020. [PMID: 32983137 DOI: 10.3389/fimmu.2020.01986/bibtex] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
Monoclonal antibodies (mAbs) have become one of the most important classes of biopharmaceutical products, and they continue to dominate the universe of biopharmaceutical markets in terms of approval and sales. They are the most profitable single product class, where they represent six of the top ten selling drugs. At the beginning of the 1990s, an in vitro antibody selection technology known as antibody phage display was developed by John McCafferty and Sir. Gregory Winter that enabled the discovery of human antibodies for diverse applications, particularly antibody-based drugs. They created combinatorial antibody libraries on filamentous phage to be utilized for generating antigen specific antibodies in a matter of weeks. Since then, more than 70 phage-derived antibodies entered clinical studies and 14 of them have been approved. These antibodies are indicated for cancer, and non-cancer medical conditions, such as inflammatory, optical, infectious, or immunological diseases. This review will illustrate the utility of phage display as a powerful platform for therapeutic antibodies discovery and describe in detail all the approved mAbs derived from phage display.
Collapse
Affiliation(s)
- Mohamed A Alfaleh
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Ahmad Bakur Mahmoud
- College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Almohanad A Alkayyal
- Department of Medical Laboratory Technology, University of Tabuk, Tabuk, Saudi Arabia
| | - Martina L Jones
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Stephen M Mahler
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Anwar M Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
226
|
Abstract
Much of our knowledge regarding the interactions between epithelial tissues and the immune system has been gathered from animal models and co-cultures with cell lines. However, unique features of human cells cannot be modelled in mice, and cell lines are often transformed or genetically immortalized. Organoid technology has emerged as a powerful tool to maintain epithelial cells in a near-native state. In this Review, we discuss how organoids are being used in immunological research to understand the role of epithelial cell-immune cell interactions in tissue development and homeostasis, as well as in diseases such as cancer.
Collapse
|
227
|
Xia X, Xia J, Yang H, Li Y, Liu S, Cao Y, Tang L, Yu X. Baicalein blocked cervical carcinoma cell proliferation by targeting CCND1 via Wnt/β-catenin signaling pathway. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2729-2736. [PMID: 31284780 DOI: 10.1080/21691401.2019.1636055] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The purpose of this study was to investigate the inhibitory effect of baicalein on the proliferation of cervical carcinoma cells and stimulate cervical carcinoma cells with baicalein. MTT method was used to observe cell proliferation. Flow cytometry was used to observe cell cycle, and gene technology was used to observe the expression of corresponding genes at the level of gene and protein. β-catenin activity was assessed using Western blot and ChIP. Baicalein suppressed cervical carcinoma cell HeLa proliferation by enhancing the activity of caspase-3. Baicalein blocked cell cycle at G0/G1 stage by inhibiting the expression of some genes. At the same time, it can prevent the nuclear translocation of β-catenin and inhibit the activity of Wnt. When the Wnt signaling pathway is increased, the proliferation of HeLa cells is inhibited, and apoptosis is promoted in this way. In conclusion, it indicated that baicalein inhibits cervical carcinoma progression by targeting CCND1 via Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Xiulian Xia
- a Department of Obstetrics and Gynecology, The Affiliated TCM Hospital of South West Medical University , Luzhou , China
| | - Jiyi Xia
- b School of Medical Information and Engineering, Southwest Medical University , Luzhou , China
| | - Hai Yang
- a Department of Obstetrics and Gynecology, The Affiliated TCM Hospital of South West Medical University , Luzhou , China
| | - Yan Li
- c Medicine Experimental Center, The Affiliated Hospital of Southwest Medical University , Luzhou , China
| | - Shengyue Liu
- a Department of Obstetrics and Gynecology, The Affiliated TCM Hospital of South West Medical University , Luzhou , China
| | - Yong Cao
- c Medicine Experimental Center, The Affiliated Hospital of Southwest Medical University , Luzhou , China
| | - Li Tang
- c Medicine Experimental Center, The Affiliated Hospital of Southwest Medical University , Luzhou , China
| | - Xiaolan Yu
- a Department of Obstetrics and Gynecology, The Affiliated TCM Hospital of South West Medical University , Luzhou , China
| |
Collapse
|
228
|
Role of the Wnt signalling pathway in the development of endothelial disorders in response to hyperglycaemia. Expert Rev Mol Med 2019; 21:e7. [PMID: 31796147 DOI: 10.1017/erm.2019.8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Diabetes mellitus (DM) is the most common metabolic disease. A WHO report from 2016 indicates that 422 million people worldwide suffer from DM or hyperglycaemia because of impaired glucose metabolism. Chronic hyperglycaemia leads to micro- and macrovessel damage, which may result in life-threatening complications. The Wnt pathway regulates cell proliferation and survival by modulating the expression of genes that control cell differentiation. Three linked Wnt pathways have been discovered thus far: a β-catenin-dependent pathway and two pathways independent of β-catenin - the planar cell polarity pathway and calcium-dependent pathway. The Wnt pathway regulates genes associated with inflammation, cell cycle, angiogenesis, fibrinolysis and other molecular processes. AREAS COVERED This review presents the current state of knowledge regarding the contribution of the Wnt pathway to endothelial ageing under hyperglycaemic conditions and provides new insights into the molecular basis of diabetic endothelial dysfunction. CONCLUSION The β-catenin-dependent pathway is a potential target in the prophylaxis and treatment of early-stage diabetes-related vascular complications. However, the underlying molecular mechanisms remain largely undetermined and require further investigation.
Collapse
|
229
|
Clara JA, Monge C, Yang Y, Takebe N. Targeting signalling pathways and the immune microenvironment of cancer stem cells - a clinical update. Nat Rev Clin Oncol 2019; 17:204-232. [PMID: 31792354 DOI: 10.1038/s41571-019-0293-2] [Citation(s) in RCA: 487] [Impact Index Per Article: 81.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2019] [Indexed: 02/06/2023]
Abstract
Cancer stem cells (CSCs) have important roles in tumour development, relapse and metastasis; the intrinsic self-renewal characteristics and tumorigenic properties of these cells provide them with unique capabilities to resist diverse forms of anticancer therapy, seed recurrent tumours, and disseminate to and colonize distant tissues. The findings of several studies indicate that CSCs originate from non-malignant stem or progenitor cells. Accordingly, inhibition of developmental signalling pathways that are crucial for stem and progenitor cell homeostasis and function, such as the Notch, WNT, Hedgehog and Hippo signalling cascades, continues to be pursued across multiple cancer types as a strategy for targeting the CSCs hypothesized to drive cancer progression - with some success in certain malignancies. In addition, with the renaissance of anticancer immunotherapy, a better understanding of the interplay between CSCs and the tumour immune microenvironment might be the key to unlocking a new era of oncological treatments associated with a reduced propensity for the development of resistance and with enhanced antimetastatic activity, thus ultimately resulting in improved patient outcomes. Herein, we provide an update on the progress to date in the clinical development of therapeutics targeting the Notch, WNT, Hedgehog and Hippo pathways. We also discuss the interactions between CSCs and the immune system, including the potential immunological effects of agents targeting CSC-associated developmental signalling pathways, and provide an overview of the emerging approaches to CSC-targeted immunotherapy.
Collapse
Affiliation(s)
- Joseph A Clara
- National Heart Lung and Blood Institute, NIH, Bethesda, MD, USA
| | - Cecilia Monge
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Yingzi Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, Dana-Farber/Harvard Cancer Center, Boston, MA, USA
| | - Naoko Takebe
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
230
|
Bats ML, Bougaran P, Peghaire C, Gueniot F, Abelanet A, Chan H, Séguy C, Jeanningros S, Jaspard-Vinassa B, Couffinhal T, Duplàa C, Dufourcq P. Therapies targeting Frizzled-7/β-catenin pathway prevent the development of pathological angiogenesis in an ischemic retinopathy model. FASEB J 2019; 34:1288-1303. [PMID: 31914666 DOI: 10.1096/fj.201901886r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 10/28/2019] [Accepted: 11/11/2019] [Indexed: 12/24/2022]
Abstract
Retinopathies remain major causes of visual impairment in diabetic patients and premature infants. Introduction of anti-angiogenic drugs targeting vascular endothelial growth factor (VEGF) has transformed therapy for these proliferative retinopathies. However, limitations associated with anti-VEGF medications require to unravel new pathways of vessel growth to identify potential drug targets. Here, we investigated the role of Wnt/Frizzled-7 (Fzd7) pathway in a mouse model of oxygen-induced retinopathy (OIR). Using transgenic mice, which enabled endothelium-specific and time-specific Fzd7 deletion, we demonstrated that Fzd7 controls both vaso-obliteration and neovascular phases (NV). Deletion of Fzd7 at P12, after the ischemic phase of OIR, prevented formation of aberrant neovessels into the vitreous by suppressing proliferation of endothelial cells (EC) in tufts. Next we validated in vitro two Frd7 blocking strategies: a monoclonal antibody (mAbFzd7) against Fzd7 and a soluble Fzd7 receptor (CRD). In vivo a single intravitreal microinjection of mAbFzd7 or CRD significantly attenuated retinal neovascularization (NV) in mice with OIR. Molecular analysis revealed that Fzd7 may act through the activation of Wnt/β-catenin and Jagged1 expression to control EC proliferation in extra-retinal neovessels. We identified Fzd7/β-catenin signaling as new regulator of pathological retinal NV. Fzd7 appears to be a potent pharmacological target to prevent or treat aberrant angiogenesis of ischemic retinopathies.
Collapse
Affiliation(s)
- Marie-Lise Bats
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France.,Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France.,Service de Biochimie clinique, CHU de Bordeaux, Bordeaux, France
| | - Pauline Bougaran
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France.,Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France
| | - Claire Peghaire
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France.,NHLI-Vascular Science, Imperial College London, London, UK
| | - Florian Gueniot
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France.,Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France
| | - Alice Abelanet
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France.,Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France
| | - Hélène Chan
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France
| | - Camille Séguy
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France
| | | | - Béatrice Jaspard-Vinassa
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France.,Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France
| | - Thierry Couffinhal
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France.,Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France.,Service des Maladies cardiaques et vasculaires, CHU de Bordeaux, Bordeaux, France
| | - Cécile Duplàa
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France.,Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France
| | - Pascale Dufourcq
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France.,Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France
| |
Collapse
|
231
|
Dhar N, Arsiwala A, Murali S, Kane RS. "Trim"ming PolyQ proteins with engineered PML. Biotechnol Bioeng 2019; 117:362-371. [PMID: 31710088 DOI: 10.1002/bit.27220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/31/2019] [Accepted: 11/03/2019] [Indexed: 01/06/2023]
Abstract
Protein abnormalities are the major cause of neurodegenerative diseases such as spinocerebellar ataxia (SCA). Protein misfolding and impaired degradation leads to the build-up of protein aggregates inside the cell, which may further cause cellular degeneration. Reducing levels of either the soluble misfolded form of the protein or its precipitated aggregate, even marginally, could significantly improve cellular health. Despite numerous pre-existing strategies to target these protein aggregates, there is considerable room to improve their specificity and efficiency. In this study, we demonstrated the enhanced intracellular degradation of both monomers and aggregates of mutant ataxin1 (Atxn1 82Q) by engineering an E3 ubiquitin ligase enzyme, promyelocytic leukemia protein (PML). Specifically, we showed enhanced degradation of both soluble and aggregated Atxn1 82Q in mammalian cells by targeting this protein using PML fused to single chain variable fragments (scFvs) specific for monomers and aggregates of the target protein. The ability to solubilize Atxn1 82Q aggregates was due to the PML-mediated enhanced SUMOylation of the target protein. This ability to reduce the intracellular levels of both misfolded forms of Atxn1 82Q may not only be useful for treating SCA, but also applicable for the treatment of other PolyQ disorders.
Collapse
Affiliation(s)
- Neha Dhar
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | - Ammar Arsiwala
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | - Shruthi Murali
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | - Ravi S Kane
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
232
|
Azbazdar Y, Ozalp O, Sezgin E, Veerapathiran S, Duncan AL, Sansom MSP, Eggeling C, Wohland T, Karaca E, Ozhan G. More Favorable Palmitic Acid Over Palmitoleic Acid Modification of Wnt3 Ensures Its Localization and Activity in Plasma Membrane Domains. Front Cell Dev Biol 2019; 7:281. [PMID: 31803740 PMCID: PMC6873803 DOI: 10.3389/fcell.2019.00281] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/31/2019] [Indexed: 12/17/2022] Open
Abstract
While the lateral organization of plasma membrane components has been shown to control binding of Wnt ligands to their receptors preferentially in the ordered membrane domains, the role of posttranslational lipid modification of Wnt on this selective binding is unknown. Here, we identify that the canonical Wnt is presumably acylated by palmitic acid, a saturated 16-carbon fatty acid, at a conserved serine residue. Acylation of Wnt3 is dispensable for its secretion and binding to Fz8 while it is essential for Wnt3's proper binding and domain-like diffusion in the ordered membrane domains. We further unravel that non-palmitoylated Wnt3 is unable to activate Wnt/β-catenin signaling either in zebrafish embryos or in mammalian cells. Based on these results, we propose that the lipidation of canonical Wnt, presumably by a saturated fatty acid, determines its competence in interacting with the receptors in the appropriate domains of the plasma membrane, ultimately keeping the signaling activity under control.
Collapse
Affiliation(s)
- Yagmur Azbazdar
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, Izmir, Turkey
| | - Ozgun Ozalp
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, Izmir, Turkey
| | - Erdinc Sezgin
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Sapthaswaran Veerapathiran
- Department of Biological Sciences and Center for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| | - Anna L. Duncan
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Mark S. P. Sansom
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Christian Eggeling
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Department of Super-Resolution Microscopy, Institute for Applied Optics and Biophysics, Friedrich-Schiller-University Jena, Jena, Germany
- Department of Biophysical Imaging, Leibniz Institute of Photonic Technology e.V., Jena, Germany
| | - Thorsten Wohland
- Department of Biological Sciences and Center for BioImaging Sciences, National University of Singapore, Singapore, Singapore
- Department of Chemistry, National University of Singapore, Singapore, Singapore
| | - Ezgi Karaca
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, Izmir, Turkey
| | - Gunes Ozhan
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, Izmir, Turkey
| |
Collapse
|
233
|
The Regulation of Bone Metabolism and Disorders by Wnt Signaling. Int J Mol Sci 2019; 20:ijms20225525. [PMID: 31698687 PMCID: PMC6888566 DOI: 10.3390/ijms20225525] [Citation(s) in RCA: 234] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/03/2019] [Accepted: 11/04/2019] [Indexed: 12/21/2022] Open
Abstract
Wnt, a secreted glycoprotein, has an approximate molecular weight of 40 kDa, and it is a cytokine involved in various biological phenomena including ontogeny, morphogenesis, carcinogenesis, and maintenance of stem cells. The Wnt signaling pathway can be classified into two main pathways: canonical and non-canonical. Of these, the canonical Wnt signaling pathway promotes osteogenesis. Sclerostin produced by osteocytes is an inhibitor of this pathway, thereby inhibiting osteogenesis. Recently, osteoporosis treatment using an anti-sclerostin therapy has been introduced. In this review, the basics of Wnt signaling, its role in bone metabolism and its involvement in skeletal disorders have been covered. Furthermore, the clinical significance and future scopes of Wnt signaling in osteoporosis, osteoarthritis, rheumatoid arthritis and neoplasia are discussed.
Collapse
|
234
|
Mirza Z, Karim S. Nanoparticles-based drug delivery and gene therapy for breast cancer: Recent advancements and future challenges. Semin Cancer Biol 2019; 69:226-237. [PMID: 31704145 DOI: 10.1016/j.semcancer.2019.10.020] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 10/15/2019] [Accepted: 10/29/2019] [Indexed: 12/24/2022]
Abstract
Breast cancer (BC) is amongst the most lethal cancer among females and conventional treatment methods like surgery, radiotherapy and chemotherapy are not effective enough as expected and suffer concerns of low bioavailability, low cellular uptake, emerging resistance, and adverse toxicities. Gene therapy using free nucleic acids has potential to deal with key candidate genes of BC, but their effect is retarded due to poor cell uptake and instability in circulation. The rapidly evolving field of nanomedicine aiming targeted drug/gene delivery curtailing BC promises to overcome the limitations of conventional therapies. Nanoparticles can be game changer for BC gene therapy as they can be effective carrier of specific drug/gene by improving the circulation time, enhancing bioavailability, reducing the immune system based recognition chances, and delivering the gene regulator accurately. Herein, we discuss the mechanism of nanoparticles targeted drug delivery, recent advancement of therapeutic strategies of nanoparticles based carriers for small interfering RNA, and microRNA, and gene augmentation therapies in BC. We also discuss the future prospect and challenges of nanoparticle-based therapies for BC.
Collapse
Affiliation(s)
- Zeenat Mirza
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Lab Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Sajjad Karim
- Department of Medical Lab Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia; Center of Excellence in Genomic Medicine Research, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
235
|
Wnt Signaling in the Regulation of Immune Cell and Cancer Therapeutics. Cells 2019; 8:cells8111380. [PMID: 31684152 PMCID: PMC6912555 DOI: 10.3390/cells8111380] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/21/2019] [Accepted: 11/01/2019] [Indexed: 12/17/2022] Open
Abstract
Wnt signaling is one of the important pathways to play a major role in various biological processes, such as embryonic stem-cell development, tissue regeneration, cell differentiation, and immune cell regulation. Recent studies suggest that Wnt signaling performs an essential function in immune cell modulation and counteracts various disorders. Nonetheless, the emerging role and mechanism of action of this signaling cascade in immune cell regulation, as well as its involvement in various cancers, remain debatable. The Wnt signaling in immune cells is very diverse, e.g., the tolerogenic role of dendritic cells, the development of natural killer cells, thymopoiesis of T cells, B-cell-driven initiation of T-cells, and macrophage actions in tissue repair, regeneration, and fibrosis. The purpose of this review is to highlight the current therapeutic targets in (and the prospects of) Wnt signaling, as well as the potential suitability of available modulators for the development of cancer immunotherapies. Although there are several Wnt inhibitors relevant to cancer, it would be worthwhile to extend this approach to immune cells.
Collapse
|
236
|
Eyre R, Alférez DG, Santiago-Gómez A, Spence K, McConnell JC, Hart C, Simões BM, Lefley D, Tulotta C, Storer J, Gurney A, Clarke N, Brown M, Howell SJ, Sims AH, Farnie G, Ottewell PD, Clarke RB. Microenvironmental IL1β promotes breast cancer metastatic colonisation in the bone via activation of Wnt signalling. Nat Commun 2019; 10:5016. [PMID: 31676788 PMCID: PMC6825219 DOI: 10.1038/s41467-019-12807-0] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 10/02/2019] [Indexed: 02/07/2023] Open
Abstract
Dissemination of tumour cells to the bone marrow is an early event in breast cancer, however cells may lie dormant for many years before bone metastases develop. Treatment for bone metastases is not curative, therefore new adjuvant therapies which prevent the colonisation of disseminated cells into metastatic lesions are required. There is evidence that cancer stem cells (CSCs) within breast tumours are capable of metastasis, but the mechanism by which these colonise bone is unknown. Here, we establish that bone marrow-derived IL1β stimulates breast cancer cell colonisation in the bone by inducing intracellular NFkB and CREB signalling in breast cancer cells, leading to autocrine Wnt signalling and CSC colony formation. Importantly, we show that inhibition of this pathway prevents both CSC colony formation in the bone environment, and bone metastasis. These findings establish that targeting IL1β-NFKB/CREB-Wnt signalling should be considered for adjuvant therapy to prevent breast cancer bone metastasis.
Collapse
Affiliation(s)
- Rachel Eyre
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - Denis G Alférez
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - Angélica Santiago-Gómez
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - Kath Spence
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - James C McConnell
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Claire Hart
- Genito Urinary Cancer Research Group, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - Bruno M Simões
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - Diane Lefley
- Academic Unit of Clinical Oncology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, S10 2RX, UK
| | - Claudia Tulotta
- Academic Unit of Clinical Oncology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, S10 2RX, UK
| | - Joanna Storer
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - Austin Gurney
- OncoMed Pharmaceuticals, Redwood City, CA, 94063, USA
| | - Noel Clarke
- Department of Urology, Salford Royal Hospital NHS Foundation Trust, Stott Lane, Salford, M6 8HD, UK
| | - Mick Brown
- Genito Urinary Cancer Research Group, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - Sacha J Howell
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
- The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - Andrew H Sims
- Applied Bioinformatics of Cancer Group, Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XR, UK
| | - Gillian Farnie
- Structural Genomics Consortium, NDORMS, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK
| | - Penelope D Ottewell
- Academic Unit of Clinical Oncology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, S10 2RX, UK.
| | - Robert B Clarke
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK.
| |
Collapse
|
237
|
Alves Pinto I, Freitas Da Silveira NJ. In Silico Identification of Potential Inhibitors of the Wnt Signaling Pathway in Human Breast Cancer. J Comput Biol 2019; 27:999-1010. [PMID: 31647315 DOI: 10.1089/cmb.2019.0311] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer is the leading worldwide cause of cancer-related deaths in women. The prospection and development of new substances with antitumoral potential is of great importance for the treatment of this disease. The objective of this work was to identify a commercial drug or ligand that could potentially bind to the FZD7 transmembrane protein and inactivate the Wnt signaling pathway in triple-negative breast cancer cells. We aimed at computationally modeling the FZD7, Wnt3, and Wnt3a proteins, at making them available in protein model databases, and at conducting docking analysis to assess the binding free energy between FZD7 and the selected ligands. The Wnt3 and Wnt3a proteins were modeled by homology modeling, and the FZD7 protein was modeled by homology modeling and ab initio modeling. The ligands were selected based on their similarity to the palmitoleic acid and were gathered from the ZINC database. A total of 30 commercially available ligands were found in the ZINC database. The docking results show that the ligands zinc08221009, zinc13546050, zinc05260769, zinc04529321, and zinc05972969 are good candidates for novel drug development. The created models and conducted analysis by this work will most certainly help in future research on the Wnt signaling pathway and its components.
Collapse
Affiliation(s)
- Icaro Alves Pinto
- Laboratory of Molecular Modeling and Computer Simulations, University of Alfenas, Alfenas, Brazil
| | | |
Collapse
|
238
|
Dzobo K, Thomford NE, Senthebane DA. Targeting the Versatile Wnt/β-Catenin Pathway in Cancer Biology and Therapeutics: From Concept to Actionable Strategy. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 23:517-538. [PMID: 31613700 DOI: 10.1089/omi.2019.0147] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This expert review offers a critical synthesis of the latest insights and approaches at targeting the Wnt/β-catenin pathway in various cancers such as colorectal cancer, melanoma, leukemia, and breast and lung cancers. Notably, from organogenesis to cancer, the Wnt/β-catenin signaling displays varied and highly versatile biological functions in animals, with virtually all tissues requiring the Wnt/β-catenin signaling in one way or the other. Aberrant expression of the members of the Wnt/β-catenin has been implicated in many pathological conditions, particularly in human cancers. Mutations in the Wnt/β-catenin pathway genes have been noted in diverse cancers. Biochemical and genetic data support the idea that inhibition of Wnt/β-catenin signaling is beneficial in cancer therapeutics. The interaction of this important pathway with other signaling systems is also noteworthy, but remains as an area for further research and discovery. In addition, formation of different complexes by components of the Wnt/β-catenin pathway and the precise roles of these complexes in the cytoplasmic milieu are yet to be fully elucidated. This article highlights the latest medical technologies in imaging, single-cell omics, use of artificial intelligence (e.g., machine learning techniques), genome sequencing, quantum computing, molecular docking, and computational softwares in modeling interactions between molecules and predicting protein-protein and compound-protein interactions pertinent to the biology and therapeutic value of the Wnt/β-catenin signaling pathway. We discuss these emerging technologies in relationship to what is currently needed to move from concept to actionable strategies in translating the Wnt/β-catenin laboratory discoveries to Wnt-targeted cancer therapies and diagnostics in the clinic.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa.,Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Nicholas Ekow Thomford
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Dimakatso A Senthebane
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa.,Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
239
|
Li N, Wei L, Liu X, Bai H, Ye Y, Li D, Li N, Baxa U, Wang Q, Lv L, Chen Y, Feng M, Lee B, Gao W, Ho M. A Frizzled-Like Cysteine-Rich Domain in Glypican-3 Mediates Wnt Binding and Regulates Hepatocellular Carcinoma Tumor Growth in Mice. Hepatology 2019; 70:1231-1245. [PMID: 30963603 PMCID: PMC6783318 DOI: 10.1002/hep.30646] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 04/02/2019] [Indexed: 12/12/2022]
Abstract
Wnt signaling is one of the key regulators of hepatocellular carcinoma (HCC) tumor progression. In addition to the classical receptor frizzled (FZD), various coreceptors including heparan sulfate proteoglycans (HSPGs) are involved in Wnt activation. Glypican-3 (GPC3) is an HSPG that is overexpressed in HCC and functions as a Wnt coreceptor that modulates HCC cell proliferation. These features make GPC3 an attractive target for liver cancer therapy. However, the precise interaction of GPC3 and Wnt and how GPC3, Wnt, and FZD cooperate with each other are poorly understood. In this study, we established a structural model of GPC3 containing a putative FZD-like cysteine-rich domain at its N-terminal lobe. We found that F41 and its surrounding residues in GPC3 formed a Wnt-binding groove that interacted with the middle region located between the lipid thumb domain and the index finger domain of Wnt3a. Mutating residues in this groove significantly inhibited Wnt3a binding, β-catenin activation, and the transcriptional activation of Wnt-dependent genes. In contrast with the heparan sulfate chains, the Wnt-binding groove that we identified in the protein core of GPC3 seemed to promote Wnt signaling in conditions when FZD was not abundant. Specifically, blocking this domain using an antibody inhibited Wnt activation. In HCC cells, mutating residue F41 on GPC3 inhibited activation of β-catenin in vitro and reduced xenograft tumor growth in nude mice compared with cells expressing wild-type GPC3. Conclusion: Our investigation demonstrates a detailed interaction of GPC3 and Wnt3a, reveals the precise mechanism of GPC3 acting as a Wnt coreceptor, and provides a potential target site on GPC3 for Wnt blocking and HCC therapy.
Collapse
Affiliation(s)
- Na Li
- Key Laboratory of Human Functional Genomics of Jiangsu Province, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Liwen Wei
- Key Laboratory of Human Functional Genomics of Jiangsu Province, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China.,Bio-medical Center, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, P.R. China
| | - Xiaoyu Liu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Hongjun Bai
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yvonne Ye
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dan Li
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.,School of Life Sciences, East China Normal University, Shanghai 200241, P.R. China
| | - Nan Li
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ulrich Baxa
- Electron Microscopy Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Qun Wang
- School of Life Sciences, East China Normal University, Shanghai 200241, P.R. China
| | - Ling Lv
- Liver Transplantation Center of the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province 210029, P.R. China
| | - Yun Chen
- Key Laboratory of Human Functional Genomics of Jiangsu Province, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Mingqian Feng
- Bio-medical Center, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, P.R. China
| | - Byungkook Lee
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Gao
- Key Laboratory of Human Functional Genomics of Jiangsu Province, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China.,Corresponding to: Dr. Wei Gao, School of Basic Medical Science, Nanjing Medical University, 101 Longmian Road, Xuehai Building, Room A110, Nanjing, Jiangsu, 211166, P.R. China. Tel: 86-25-86869471; Fax: 86-25-86869471, . Dr. Mitchell Ho, Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Room 5002, Bethesda, MD 20892-4264. Tel: (240)760-7848; Fax: (301)402-1344;
| | - Mitchell Ho
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.,Corresponding to: Dr. Wei Gao, School of Basic Medical Science, Nanjing Medical University, 101 Longmian Road, Xuehai Building, Room A110, Nanjing, Jiangsu, 211166, P.R. China. Tel: 86-25-86869471; Fax: 86-25-86869471, . Dr. Mitchell Ho, Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Room 5002, Bethesda, MD 20892-4264. Tel: (240)760-7848; Fax: (301)402-1344;
| |
Collapse
|
240
|
Tang T, Guo C, Xia T, Zhang R, Zen K, Pan Y, Jin L. LncCCAT1 Promotes Breast Cancer Stem Cell Function through Activating WNT/β-catenin Signaling. Theranostics 2019; 9:7384-7402. [PMID: 31695775 PMCID: PMC6831302 DOI: 10.7150/thno.37892] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 08/05/2019] [Indexed: 02/07/2023] Open
Abstract
Background: Breast cancer stem cells (BCSCs) play an essential role in facilitating breast cancer relapse and metastasis. The underlying mechanism, however, remains incompletely understood. In the current study, we investigated the clinical significance, biological function and mechanism of a long noncoding RNA CCAT1 (LncCCAT1) in BCSCs. Methods: Firstly, lncRNAs expression in poorly differentiated breast cancer tissues and BCSCs were measured by lncRNA microarray and confirmed in breast cancer tissues and cell lines. The functional roles and mechanisms of LncCCAT1 were further investigated by gain and loss of function assays in vitro and in vivo. Results: LncCCAT1 is markedly upregulated in breast cancer tissues BCSCs and is correlated with poor outcomes in breast cancer patients. Overexpression of LncCCAT1 contributes to the proliferation, stemness, migration and invasion capacities of BCSCs. Mechanistic investigation suggests that LncCCAT1 can interact with miR-204/211, miR-148a/152 and Annexin A2(ANXA2), then upregulate T-cell factor 4 (TCF4) or promote translocation of β-catenin to the nucleus where it activates TCF4, leading to the activation of wingless/integrated (Wnt) signaling. Furthermore, TCF4 can also bind to the promoter of LncCCAT1 to promote LncCCAT1 transcription, thus forming a positive feedback regulatory circuit of LncCCAT1-TCF4-LncCCAT1 in BCSCs. Conclusions: LncCCAT1 plays an important role in breast cancer progression and may serve as a novel target for breast cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Tingting Tang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University. 24 Tongjiaxiang Avenue, Nanjing, Jiangsu, China
| | - Changying Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University. 24 Tongjiaxiang Avenue, Nanjing, Jiangsu, China
| | - Tiansong Xia
- Department of Breast Surgery, Breast Disease Center of Jiangsu Province, First Affiliated Hospital of Nanjing Medical University. 300 Guangzhou Road, Nanjing, Jiangsu province, China
| | - Rui Zhang
- Department of Biochemistry and Molecular Biology and Key Laboratory of Breast Cancer Prevention and Treatment, Ministry of Education, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ke Zen
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu province, China
| | - Yi Pan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University. 24 Tongjiaxiang Avenue, Nanjing, Jiangsu, China
| | - Liang Jin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University. 24 Tongjiaxiang Avenue, Nanjing, Jiangsu, China
| |
Collapse
|
241
|
Li X, Xiang Y, Li F, Yin C, Li B, Ke X. WNT/β-Catenin Signaling Pathway Regulating T Cell-Inflammation in the Tumor Microenvironment. Front Immunol 2019; 10:2293. [PMID: 31616443 PMCID: PMC6775198 DOI: 10.3389/fimmu.2019.02293] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 09/11/2019] [Indexed: 01/26/2023] Open
Abstract
Immunotherapy with checkpoint inhibitors has greatly prolonged the overall survival of cancer patients in melanoma and many other cancer types. However, only a subset of patients shows clinical responses from these interventions, which was predicated by the T cell-inflamed tumor microenvironment. T cell-inflamed phenotype is characterized by the infiltration of CD8+ T cells, CD8α/CD103-lineage dendritic cells (DCs), as well as high density of forkhead box P3 (FoxP3)+ regulatory T cells (Tregs) that are associated with the efficacy of immune checkpoint blockade. A number of regulators has been associated with T cell-inflammation in the tumor microenvironment, and WNT/β-catenin signaling is one of the best characterized. The tumor-intrinsic WNT/β-catenin signaling activation is frequently associated with poor spontaneous T cell infiltration across most human cancers. In this article, we review the essential roles of WNT/β-catenin signaling in the T cell-inflamed and non-T cell-inflamed tumor microenvironment, including the development and function of immune cells, activation of immune exclusion of tumor cells, and cancer immunosurveillance. We also discuss the impact of this pathway in driving the non-T cell-inflamed tumor microenvironment in other tumor types. To improve immunotherapy efficacy, we argue that targeting Wnt/β-catenin signaling should be a high priority for combinational cancer therapy to restore T cell infiltration.
Collapse
Affiliation(s)
- Xin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Yanwei Xiang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fulun Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chengqian Yin
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Bin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Xisong Ke
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Center for Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
242
|
Cosin-Roger J, Ortiz-Masià MD, Barrachina MD. Macrophages as an Emerging Source of Wnt Ligands: Relevance in Mucosal Integrity. Front Immunol 2019; 10:2297. [PMID: 31608072 PMCID: PMC6769121 DOI: 10.3389/fimmu.2019.02297] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023] Open
Abstract
The Wnt signaling pathway is a conserved pathway involved in important cellular processes such as the control of embryonic development, cellular polarity, cellular migration, and cell proliferation. In addition to playing a central role during embryogenesis, this pathway is also an essential part of adult homeostasis. Indeed, it controls the proliferation of epithelial cells in different organs such as intestine, lung, and kidney, and guarantees the maintenance of the mucosa in physiological conditions. The origin of this molecular pathway is the binding between Wnt ligands (belonging to a family of 19 different homologous secreted glycoproteins) and their specific membrane receptors, from the Frizzled receptor family. This specific interaction triggers the activation of the signaling cascade, which in turn activates or suppresses the expression of different genes in order to change the behavior of the cell. On the other hand, alterations of this pathway have been described in pathological conditions such as inflammation, fibrosis, and cancer. In recent years, macrophages-among other cell types-have emerged as a potential source of Wnt ligands. Due to their high plasticity, macrophages, which are central to the innate immune response, are capable of adopting different phenotypes depending on their microenvironment. In the past, two different phenotypes were described: a proinflammatory phenotype-M1 macrophages-and an anti-inflammatory phenotype-M2 macrophages-and a selective expression of Wnt ligands has been associated with said phenotypes. However, nowadays it is assumed that macrophages in vivo move through a continual spectrum of functional phenotypes. In both physiological and pathological (inflammation, fibrosis and cancer) conditions, the accumulation and polarization of macrophages conditions the future of the tissue, facilitating various scenarios, such as resolution of inflammation, activation of fibrosis, and cancer development due to the modulation of the Wnt signaling pathway, in autocrine and paracrine manner. In this work, we provide an overview of studies that have explored the role of macrophages and how they act as a source of Wnt ligands and as mediators of mucosal integrity.
Collapse
Affiliation(s)
| | - Mª Dolores Ortiz-Masià
- Departamento de Medicina, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Mª Dolores Barrachina
- Departamento de Farmacología and CIBER, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| |
Collapse
|
243
|
Liao YM, Song Y, Li YK, Du JH, Zhou Y. SOX17, β-catenin and CyclinD1 expression in the endometrioid adenocarcinoma and influence of 5-AZA on expression. Cancer Gene Ther 2019; 27:256-263. [PMID: 31543512 DOI: 10.1038/s41417-019-0135-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/12/2019] [Accepted: 07/20/2019] [Indexed: 12/25/2022]
Abstract
The present study discusses the expression and effect of the SOX17 gene in endometrioid adenocarcinoma. MTT assay is performed to determine the growth inhibition ratio of the DNA methyltransferase inhibitor 5-AZA for endometrial carcinoma cells, and the real-time fluorescence quantification PCR (qRT-PCR) was used to detect the mRNA expression of SOX17, β-catenin, and CyclinD1 in endometrial carcinoma tissues before and after using 5-AZA to treat the endometrial carcinoma cell line. There were 30 cases on endometrioid adenocarcinoma tissues and 10 cases on normal endometrial tissues. The results revealed that the expression of SOX17 in endometrioid adenocarcinoma tissues was downregulated (P < 0.05), the expression of β-catenin and CyclinD1 was upregulated (P < 0.05), and the expression of SOX17, CyclinD1, and β-catenin was negatively correlated (r = -0.353, P > 0.05; R = -0.463, P < 0.05). The higher the histological grade and FIGO staging were, the lower the expression level of SOX17 was (P < 0.05). After HEC1A cells were treated by 5-AZA, the cell growth inhibition was most obvious (IC50 = 12.033) at 72 h, as determined by MTT assay. After cell treatment by 5-AZA, the genetic expression of SOX17 significantly increased, when compared with that before treatment (P < 0.05), while the genetic expression of β-catenin and CyclinD1 significantly declined (P < 0.05). These results indicate that the expression level of SOX17 in endometrioid adenocarcinoma declined, and the upregulated expression level of SOX17 in cells inhibited the growth of tumor cells.
Collapse
Affiliation(s)
- Yu-Mei Liao
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Yang Song
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuan-Kun Li
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jie-Hua Du
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying Zhou
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
244
|
Hansen S, Nile AH, Mehta SC, Fuhrmann J, Hannoush RN. Lead Optimization Yields High Affinity Frizzled 7-Targeting Peptides That Modulate Clostridium difficile Toxin B Pathogenicity in Epithelial Cells. J Med Chem 2019; 62:7739-7750. [PMID: 31429553 DOI: 10.1021/acs.jmedchem.9b00500] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Frizzled 7 (FZD7) receptors have been shown to play a central role in intestinal stem cell regeneration and, more recently, in Clostridium difficile pathogenesis. Yet, targeting FZD7 receptors with small ligands has not been explored as an approach to block C. difficile pathogenesis. Here, we report the discovery of high affinity peptides that selectively bind to FZD7 receptors. We describe an integrated approach for lead optimization, utilizing structure-based rational design and directed evolution, to enhance the peptide binding affinity while still maintaining FZD7 receptor selectivity. This work yielded new peptide leads with picomolar binding constants to FZD7 as measured by biophysical methods. The new peptides block the interaction between C. difficile toxin B (TcdB) and FZD receptors and perturb C. difficile pathogenesis in epithelial cells. As such, our findings provide a proof of concept that targeting FZD receptors could be a viable pharmacological approach to protect epithelial cells from TcdB pathogenicity.
Collapse
|
245
|
Ram Makena M, Gatla H, Verlekar D, Sukhavasi S, K Pandey M, C Pramanik K. Wnt/β-Catenin Signaling: The Culprit in Pancreatic Carcinogenesis and Therapeutic Resistance. Int J Mol Sci 2019; 20:E4242. [PMID: 31480221 PMCID: PMC6747343 DOI: 10.3390/ijms20174242] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/22/2019] [Accepted: 08/27/2019] [Indexed: 12/24/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is responsible for 7.3% of all cancer deaths. Even though there is a steady increase in patient survival for most cancers over the decades, the patient survival rate for pancreatic cancer remains low with current therapeutic strategies. The Wnt/β-catenin pathway controls the maintenance of somatic stem cells in many tissues and organs and is implicated in pancreatic carcinogenesis by regulating cell cycle progression, apoptosis, epithelial-mesenchymal transition (EMT), angiogenesis, stemness, tumor immune microenvironment, etc. Further, dysregulated Wnt has been shown to cause drug resistance in pancreatic cancer. Although different Wnt antagonists are effective in pancreatic patients, limitations remain that must be overcome to increase the survival benefits associated with this emerging therapy. In this review, we have summarized the role of Wnt signaling in pancreatic cancer and suggested future directions to enhance the survival of pancreatic cancer patients.
Collapse
Affiliation(s)
- Monish Ram Makena
- Department of Physiology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Himavanth Gatla
- Department of Pediatric Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Dattesh Verlekar
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Sahithi Sukhavasi
- Center for Distance Learning, GITAM University, Visakhapatnam 530045, India
| | - Manoj K Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Kartick C Pramanik
- Department of Basic Sciences, Kentucky College of Osteopathic Medicine, University of Pikeville, Pikeville, KY 41501, USA.
| |
Collapse
|
246
|
Song Z, Wang H, Zhang S. Negative regulators of Wnt signaling in non-small cell lung cancer: Theoretical basis and therapeutic potency. Biomed Pharmacother 2019; 118:109336. [PMID: 31545260 DOI: 10.1016/j.biopha.2019.109336] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 07/20/2019] [Accepted: 08/05/2019] [Indexed: 02/05/2023] Open
Abstract
Significant advances in the treatment of non-small cell lung cancer (NSCLC) have been made over the past decade, and they predominantly involve molecular targets such as epidermal growth factor receptor (EGFR) mutations and anaplastic lymphoma kinase (ALK) rearrangements. However, despite the initial good response, drug resistance eventually develops. The Wnt signaling pathway has recently been considered important in embryonic development and tumorigenesis in many cancers, particularly NSCLC. Moreover, the aberrant Wnt pathway plays a significant role in NSCLC and is associated with cancer cell proliferation, metastasis, invasion and drug resistance, and the suppression of canonical or noncanonical Wnt signaling through various biological or pharmacological negative regulators has been proven to produce specific anticancer effects. Thus, blocking the Wnt pathway via its negative regulators may overcome the resistance of current treatment methods and lead to new treatment strategies for NSCLC. Therefore, in this review, we summarize recent studies on the role of negative regulators in Wnt signaling in NSCLC and the therapeutic potency of these molecules as agents and targets for NSCLC treatments.
Collapse
Affiliation(s)
- Zikuan Song
- West China School of Basic Medical Science and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Haoyu Wang
- West China School of Basic Medical Science and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shuang Zhang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
247
|
Abstract
Developmental signaling pathways control a vast array of biological processes during embryogenesis and in adult life. The WNT pathway was discovered simultaneously in cancer and development. Recent advances have expanded the role of WNT to a wide range of pathologies in humans. Here, we discuss the WNT pathway and its role in human disease and some of the advances in WNT-related treatments.
Collapse
|
248
|
Xie W, Zhang Y, Zhang S, Wang F, Zhang K, Huang Y, Zhou Z, Huang G, Wang J. Oxymatrine enhanced anti-tumor effects of Bevacizumab against triple-negative breast cancer via abating Wnt/β-Catenin signaling pathway. Am J Cancer Res 2019; 9:1796-1814. [PMID: 31497360 PMCID: PMC6726986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 07/11/2019] [Indexed: 06/10/2023] Open
Abstract
Bevacizumab, a monoclonal antibody targeting vascular endothelial growth factor A (VEGF-A), was used in combination with traditional chemotherapy as the first line treatment for metastatic colorectal cancer (mCRC), non-small cell lung cancer (NSCLC) and advanced ovarian cancer. However, it shows limited efficacy for human triple-negative breast cancer (TNBC). Bevacizumab shows potent anti-angiogenesis activity, meanwhile, it also increases invasive and metastatic properties of TNBC cells by activiting Wnt/β-Catenin pathway. To overcome this problem, and fully utilize its potency against cancer, further synergistic strategy is recommended to be developed, especially the concurrent use with those Wnt-targeting agents. Here, by screening a small library of traditional Chinese medicine, we identified a Chinese herb derived Oxymatrine, which could target Wnt/β-Catenin signaling and compromise the oncogenic effects of Bevacizumab. Bevacizumab was validated to induce epithelial-mesenchymal cell transformation (EMT) and cancer stem-like properties of TNBC cells in hypoxia/nutritional stress environment. On the contrary, Oxymatrine reversed the EMT phenotype and depleted the subpopulation of TNBC stem cells induced by Bevacizumab. Oxymatrine enhanced the anti-tumor effects of Bevacizumab in vivo, and holded the potential of reducing the risk of relapse and metastasis by impairing the self-renewal ability of TNBC stem cells. The underlying mechanism was elucidated: Bevacizumab stimulated Wnt/β-Catenin signaling pathway, and Oxymatrine could compromise this effect. On this foundation, factoring into the satisfactory anti-angiogenic activity and low toxicity, Oxymatrine is a good candidate for the synergistic therapy together with Bevacizumab for the treatment of TNBC.
Collapse
Affiliation(s)
- Wei Xie
- School of Pharmacy, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
| | - Yan Zhang
- School of Pharmacy, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
- Shanghai University of Traditional Chinese Medicine Graduate SchoolShanghai 201203, P. R. China
| | - Shiwei Zhang
- School of Pharmacy, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
| | - Fengxian Wang
- School of Pharmacy, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
- Shanghai University of Traditional Chinese Medicine Graduate SchoolShanghai 201203, P. R. China
| | - Kunchi Zhang
- School of Pharmacy, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
| | - Yanjuan Huang
- School of Pharmacy, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
| | - Zhaoli Zhou
- School of Pharmacy, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
| | - Gang Huang
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
| | - Jin Wang
- School of Pharmacy, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
| |
Collapse
|
249
|
Chen T, Lin J, Tang D, Zhang M, Wen F, Xue D, Zhang H. Paris saponin H suppresses human hepatocellular carcinoma (HCC) by inactivation of Wnt/β-catenin pathway in vitro and in vivo. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:2875-2886. [PMID: 31934124 PMCID: PMC6949715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 02/21/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death in the world. Paris polyphylla, also known as Chong-lou in China, is traditionally used as an anticancer medicine. Paris saponin H (Ps H) has been reported to be one potential antitumor active component from Paris polyphylla and shows cytotoxicity on tumor cells. However, the role of Ps H in HCC is not clear. METHODS PLC/PRF/5 and Huh7 cells were exposed to Ps H. Cell viability, migration, and invasion were measured with CCK-8 assay, EMT and Transwell assay, respectively. Western blot was employed to detect the expression of cleaved caspase 3, E-cadherin, vimentin, β-catenin, p-GSK-3β and GSK-3β. Apoptosis was assessed by flow cytometry, and caspase 3 activity assay. For in vivo experiments, xenograft tumors were induced with PLC/PRF/5 cells. RESULTS Ps H reduced cell viability and induced apoptosis in HCC cells in the dose-dependent manner; EMT and invasion were inhibited by Ps H. Ps H downregulated expression of β-catenin and p-GSK-3β; in addition, β-catenin silencing mediated Ps H-induced suppression of cell progression in PLC/PRF/5 cells. An administration of Ps H effectively suppressed the tumor growth in the HCC xenograft model in vivo. CONCLUSION Ps H suppresses HCC cell progression through downregulation of β-catenin in vitro, and inhibits xenograft tumor growth, suggesting Ps H is an attractive candidate for clinical therapy for HCC.
Collapse
Affiliation(s)
- Tiezhu Chen
- Sichuan Provincial Key Laboratory of Quality and Innovation Research of Chinese Materia Medica, Sichuan Academy of Chinese Medicine SciencesChengdu, Sichuan, China
| | - Juan Lin
- Sichuan Provincial Key Laboratory of Quality and Innovation Research of Chinese Materia Medica, Sichuan Academy of Chinese Medicine SciencesChengdu, Sichuan, China
| | - Daxuan Tang
- Sichuan Provincial Key Laboratory of Quality and Innovation Research of Chinese Materia Medica, Sichuan Academy of Chinese Medicine SciencesChengdu, Sichuan, China
| | - Mei Zhang
- Sichuan Provincial Key Laboratory of Quality and Innovation Research of Chinese Materia Medica, Sichuan Academy of Chinese Medicine SciencesChengdu, Sichuan, China
| | - Feiyan Wen
- West China School of Pharmacy, Sichuan UniversityChengdu, Sichuan, China
| | - Dan Xue
- West China School of Pharmacy, Sichuan UniversityChengdu, Sichuan, China
| | - Hao Zhang
- West China School of Pharmacy, Sichuan UniversityChengdu, Sichuan, China
| |
Collapse
|
250
|
Du FY, Zhou QF, Sun WJ, Chen GL. Targeting cancer stem cells in drug discovery: Current state and future perspectives. World J Stem Cells 2019; 11:398-420. [PMID: 31396368 PMCID: PMC6682504 DOI: 10.4252/wjsc.v11.i7.398] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/18/2019] [Accepted: 06/27/2019] [Indexed: 02/06/2023] Open
Abstract
In recent decades, cancer stem cells (CSCs) have been increasingly identified in many malignancies. CSC-related signaling pathways and their functions provide new strategies for treating cancer. The aberrant activation of related signaling pathways (e.g., Wnt, Notch, and Hedgehog pathways) has been linked to multiple types of malignant tumors, which makes these pathways attractive targets for cancer therapy. CSCs display many characteristic features, such as self-renewal, differentiation, high tumorigenicity, and drug resistance. Therefore, there is an urgent need to develop new therapeutic strategies to target these pathways to control stem cell replication, survival, and differentiation. Notable crosstalk occurs among different signaling pathways and potentially leads to compensatory escape. Therefore, multitarget inhibitors will be one of the main methods to overcome the drug resistance of CSCs. Many small molecule inhibitors of components of signaling pathways in CSCs have entered clinical trials, and some inhibitors, such as vismodegib, sonidegib, and glasdegib, have been approved. Tumor cells are susceptible to sonidegib and vismodegib resistance due to mutations in the Smo protein. The signal transducers and activators of transcription 3 (STAT3) inhibitor BBI608 is being evaluated in a phase III trial for a variety of cancers. Structural derivatives of BBI608 are the main focus of STAT3 inhibitor development, which is another strategy for CSC therapy. In addition to the potential pharmacological inhibitors targeting CSC-related signaling pathways, other methods of targeting CSCs are available, such as nano-drug delivery systems, mitochondrion targeting, autophagy, hyperthermia, immunotherapy, and CSC microenvironment targeting. In addition, we summarize the latest advances in the clinical development of agents targeting CSC-related signaling pathways and other methods of targeting CSCs.
Collapse
Affiliation(s)
- Fang-Yu Du
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning Province, China
| | - Qi-Fan Zhou
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning Province, China
| | - Wen-Jiao Sun
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning Province, China
| | - Guo-Liang Chen
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning Province, China
| |
Collapse
|