1
|
Feng M, Santhanam RK, Xing H, Zhou M, Jia H. Inhibition of γ-secretase/Notch pathway as a potential therapy for reversing cancer drug resistance. Biochem Pharmacol 2024; 220:115991. [PMID: 38135129 DOI: 10.1016/j.bcp.2023.115991] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/30/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023]
Abstract
The mechanism of tumor drug resistance is complex and may involve stem cell maintenance, epithelial-mesenchymal transition, the activation of survival signaling pathways, transporter protein expression, and tumor microenvironment remodeling, all of which are linked to γ-secretase/Notch signaling. Increasing evidence has shown that the activation of the γ-secretase/Notch pathway is a key driver of cancer progression and drug resistance development and that γ-secretase inhibitors (GSIs) may be the most promising agents for reversing chemotherapy resistance of tumors by targeting the γ-secretase/Notch pathway. Here, we systematically summarize the roles in supporting γ-secretase/Notch activation-associated transformation of cancer cells into cancer stem cells, promotion of the EMT process, PI3K/Akt, MEK/ERK and NF-κB activation, enhancement of ABC transporter protein expression, and TME alteration in mediating tumor drug resistance. Subsequently, we analyze the mechanism of GSIs targeting the γ-secretase/Notch pathway to reverse tumor drug resistance and propose the outstanding advantages of GSIs in treating breast cancer drug resistance over other tumors. Finally, we emphasize that the development of GSIs for reversing tumor drug resistance is promising.
Collapse
Affiliation(s)
- Mei Feng
- Science and Experimental Research Center of Shenyang Medical College, Shenyang 110034, China; Shenyang Key Laboratory of Vascular Biology, Shenyang 110034, China
| | - Ramesh Kumar Santhanam
- Faculty of Science and Marine Environment, University Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
| | - Huan Xing
- Science and Experimental Research Center of Shenyang Medical College, Shenyang 110034, China
| | - Mingsheng Zhou
- Science and Experimental Research Center of Shenyang Medical College, Shenyang 110034, China; Shenyang Key Laboratory of Vascular Biology, Shenyang 110034, China.
| | - Hui Jia
- School of Traditional Chinese Medicine, Shenyang Medical College, Shenyang 110034, China.
| |
Collapse
|
2
|
Yu X, He T, Tong Z, Liao L, Huang S, Fakhouri WD, Edwards DP, Xu J. Molecular mechanisms of TWIST1-regulated transcription in EMT and cancer metastasis. EMBO Rep 2023; 24:e56902. [PMID: 37680145 PMCID: PMC10626429 DOI: 10.15252/embr.202356902] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
TWIST1 induces epithelial-to-mesenchymal transition (EMT) to drive cancer metastasis. It is yet unclear what determines TWIST1 functions to activate or repress transcription. We found that the TWIST1 N-terminus antagonizes TWIST1-regulated gene expression, cancer growth and metastasis. TWIST1 interacts with both the NuRD complex and the NuA4/TIP60 complex (TIP60-Com) via its N-terminus. Non-acetylated TWIST1-K73/76 selectively interacts with and recruits NuRD to repress epithelial target gene transcription. Diacetylated TWIST1-acK73/76 binds BRD8, a component of TIP60-Com that also binds histone H4-acK5/8, to recruit TIP60-Com to activate mesenchymal target genes and MYC. Knockdown of BRD8 abolishes TWIST1 and TIP60-Com interaction and TIP60-Com recruitment to TWIST1-activated genes, resulting in decreasing TWIST1-activated target gene expression and cancer metastasis. Both TWIST1/NuRD and TWIST1/TIP60-Com complexes are required for TWIST1 to promote EMT, proliferation, and metastasis at full capacity. Therefore, the diacetylation status of TWIST1-K73/76 dictates whether TWIST1 interacts either with NuRD to repress epithelial genes, or with TIP60-Com to activate mesenchymal genes and MYC. Since BRD8 is essential for TWIST1-acK73/76 and TIP60-Com interaction, targeting BRD8 could be a means to inhibit TWIST1-activated gene expression.
Collapse
Affiliation(s)
- Xiaobin Yu
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
| | - Tao He
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
- Present address:
Institute for Cancer MedicineSouthwest Medical UniversitySichuanChina
| | - Zhangwei Tong
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
| | - Lan Liao
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
- Dan L. Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTXUSA
| | - Shixia Huang
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
- Dan L. Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTXUSA
| | - Walid D Fakhouri
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of DentistryUniversity of Texas Health Science Center at HoustonHoustonTXUSA
| | - Dean P Edwards
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
- Dan L. Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTXUSA
| | - Jianming Xu
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
- Dan L. Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTXUSA
| |
Collapse
|
3
|
Kasimanickam VR, Kasimanickam RK. Differentially Expressed Candidate miRNAs of Day 16 Bovine Embryos on the Regulation of Pregnancy Establishment in Dairy Cows. Animals (Basel) 2023; 13:3052. [PMID: 37835658 PMCID: PMC10571895 DOI: 10.3390/ani13193052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Recent advances in high-throughput in silico techniques translate experimental data into meaningful biological networks through which the role of individual proteins, interactions, and their biological functions are comprehended. The study objective was to identify differentially expressed (DE) miRNAs between the day 16 competent, elongated embryo from normal cows and the day 16 noncompetent, tubular embryos from repeat breeder cows, assimilate DE-miRNAs to their target genes, and group target genes based on biological function using in silico methods. The 84 prioritized bovine-specific miRNAs were investigated by RT-PCR, and the results showed that 19 were differentially expressed (11 up- and 8 down-regulated) in the competent embryos compared to noncompetent ones (p ≤ 0.05; fold regulation ≥ 2 magnitudes). Top-ranked integrated genes of DE-miRNAs predicted various biological and molecular functions, cellular processes, and signaling pathways. Further, analysis of the categorized groups of genes showed association with signaling pathways, turning on or off key genes and transcription factors regulating the development of embryo, placenta, and various organs. In conclusion, highly DE-miRNAs in day 16 bovine conceptus regulated the embryogenesis and pregnancy establishment. The elucidated miRNA-mRNA interactions in this study were mostly based on predictions from public databases. Therefore, the causal regulations of these interactions and mechanisms require further functional characterization.
Collapse
Affiliation(s)
- Vanmathy R. Kasimanickam
- Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA;
- AARVEE Animal Biotech LLC, Corvallis, OR 97333, USA
| | - Ramanathan K. Kasimanickam
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
4
|
Jung E, Lee YH, Ou S, Kim TY, Shin SY. EGR1 Regulation of Vasculogenic Mimicry in the MDA-MB-231 Triple-Negative Breast Cancer Cell Line through the Upregulation of KLF4 Expression. Int J Mol Sci 2023; 24:14375. [PMID: 37762678 PMCID: PMC10532327 DOI: 10.3390/ijms241814375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/09/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
Vasculogenic mimicry (VM) is an intriguing phenomenon observed in tumor masses, in which cancer cells organize themselves into capillary-like channels that closely resemble the structure and function of blood vessels. Although VM is believed to contribute to alternative tumor vascularization, the detailed regulatory mechanisms controlling these cellular processes remain poorly understood. Our study aimed to investigate the role of Early Growth Response 1 (EGR1) in regulating VM in aggressive cancer cells, specifically MDA-MB-231 triple-negative breast cancer cells. Our study revealed that EGR1 promotes the formation of capillary-like tubes by MDA-MB-231 cells in a 3-dimensional Matrigel matrix. EGR1 was observed to upregulate Kruppel-like factor 4 (KLF4) expression, which regulates the formation of the capillary-like tube structure. Additionally, our findings highlight the involvement of the ERK1/2 and p38 mitogen-activated protein kinase pathways in mediating the expression of EGR1 and KLF4, underscoring their crucial role in VM in MDA-MB-231 cells. Understanding these regulatory mechanisms will provide valuable insights into potential therapeutic targets for preventing VM during the treatment of triple-negative breast cancer.
Collapse
Affiliation(s)
- Euitaek Jung
- Department of Biological Sciences, Sanghuh College of Life Science, Konkuk University, Seoul 05029, Republic of Korea; (E.J.); (Y.H.L.); (S.O.); (T.Y.K.)
| | - Young Han Lee
- Department of Biological Sciences, Sanghuh College of Life Science, Konkuk University, Seoul 05029, Republic of Korea; (E.J.); (Y.H.L.); (S.O.); (T.Y.K.)
- Cancer and Metabolism Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Sukjin Ou
- Department of Biological Sciences, Sanghuh College of Life Science, Konkuk University, Seoul 05029, Republic of Korea; (E.J.); (Y.H.L.); (S.O.); (T.Y.K.)
| | - Tae Yoon Kim
- Department of Biological Sciences, Sanghuh College of Life Science, Konkuk University, Seoul 05029, Republic of Korea; (E.J.); (Y.H.L.); (S.O.); (T.Y.K.)
| | - Soon Young Shin
- Department of Biological Sciences, Sanghuh College of Life Science, Konkuk University, Seoul 05029, Republic of Korea; (E.J.); (Y.H.L.); (S.O.); (T.Y.K.)
- Cancer and Metabolism Institute, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
5
|
An L, Li M, Jia Q. Mechanisms of radiotherapy resistance and radiosensitization strategies for esophageal squamous cell carcinoma. Mol Cancer 2023; 22:140. [PMID: 37598158 PMCID: PMC10439611 DOI: 10.1186/s12943-023-01839-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/02/2023] [Indexed: 08/21/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the sixth most common cause of cancer-related mortality worldwide, with more than half of them occurred in China. Radiotherapy (RT) has been widely used for treating ESCC. However, radiation-induced DNA damage response (DDR) can promote the release of cytokines and chemokines, and triggers inflammatory reactions and changes in the tumor microenvironment (TME), thereby inhibiting the immune function and causing the invasion and metastasis of ESCC. Radioresistance is the major cause of disease progression and mortality in cancer, and it is associated with heterogeneity. Therefore, a better understanding of the radioresistance mechanisms may generate more reversal strategies to improve the cure rates and survival periods of ESCC patients. We mainly summarized the possible mechanisms of radioresistance in order to reveal new targets for ESCC therapy. Then we summarized and compared the current strategies to reverse radioresistance.
Collapse
Affiliation(s)
- Lingbo An
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- College of Medical Technology, Xi'an Medical University, Xi'an, China
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China.
| | - Qingge Jia
- Department of Reproductive Medicine, Xi'an International Medical Center Hospital, Northwest University, Xi'an, China.
| |
Collapse
|
6
|
Zhang C, Guo ZF, Liu W, Kazama K, Hu L, Sun X, Wang L, Lee H, Lu L, Yang XF, Summer R, Sun J. PIMT is a novel and potent suppressor of endothelial activation. eLife 2023; 12:e85754. [PMID: 37070640 PMCID: PMC10112892 DOI: 10.7554/elife.85754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/02/2023] [Indexed: 04/19/2023] Open
Abstract
Proinflammatory agonists provoke the expression of cell surface adhesion molecules on endothelium in order to facilitate leukocyte infiltration into tissues. Rigorous control over this process is important to prevent unwanted inflammation and organ damage. Protein L-isoaspartyl O-methyltransferase (PIMT) converts isoaspartyl residues to conventional methylated forms in cells undergoing stress-induced protein damage. The purpose of this study was to determine the role of PIMT in vascular homeostasis. PIMT is abundantly expressed in mouse lung endothelium and PIMT deficiency in mice exacerbated pulmonary inflammation and vascular leakage to LPS(lipopolysaccharide). Furthermore, we found that PIMT inhibited LPS-induced toll-like receptor signaling through its interaction with TNF receptor-associated factor 6 (TRAF6) and its ability to methylate asparagine residues in the coiled-coil domain. This interaction was found to inhibit TRAF6 oligomerization and autoubiquitination, which prevented NF-κB transactivation and subsequent expression of endothelial adhesion molecules. Separately, PIMT also suppressed ICAM-1 expression by inhibiting its N-glycosylation, causing effects on protein stability that ultimately translated into reduced EC(endothelial cell)-leukocyte interactions. Our study has identified PIMT as a novel and potent suppressor of endothelial activation. Taken together, these findings suggest that therapeutic targeting of PIMT may be effective in limiting organ injury in inflammatory vascular diseases.
Collapse
Affiliation(s)
- Chen Zhang
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Zhi-Fu Guo
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Wennan Liu
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Kyosuke Kazama
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Louis Hu
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Xiaobo Sun
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Lu Wang
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Hyoungjoo Lee
- Quantitative Proteomics Resource Center, University of PennsylvaniaPhiladelphiaUnited States
| | - Lin Lu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiao-Feng Yang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple UniversityPhiladelphiaUnited States
| | - Ross Summer
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Jianxin Sun
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| |
Collapse
|
7
|
Wälchli T, Bisschop J, Carmeliet P, Zadeh G, Monnier PP, De Bock K, Radovanovic I. Shaping the brain vasculature in development and disease in the single-cell era. Nat Rev Neurosci 2023; 24:271-298. [PMID: 36941369 PMCID: PMC10026800 DOI: 10.1038/s41583-023-00684-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2023] [Indexed: 03/23/2023]
Abstract
The CNS critically relies on the formation and proper function of its vasculature during development, adult homeostasis and disease. Angiogenesis - the formation of new blood vessels - is highly active during brain development, enters almost complete quiescence in the healthy adult brain and is reactivated in vascular-dependent brain pathologies such as brain vascular malformations and brain tumours. Despite major advances in the understanding of the cellular and molecular mechanisms driving angiogenesis in peripheral tissues, developmental signalling pathways orchestrating angiogenic processes in the healthy and the diseased CNS remain incompletely understood. Molecular signalling pathways of the 'neurovascular link' defining common mechanisms of nerve and vessel wiring have emerged as crucial regulators of peripheral vascular growth, but their relevance for angiogenesis in brain development and disease remains largely unexplored. Here we review the current knowledge of general and CNS-specific mechanisms of angiogenesis during brain development and in brain vascular malformations and brain tumours, including how key molecular signalling pathways are reactivated in vascular-dependent diseases. We also discuss how these topics can be studied in the single-cell multi-omics era.
Collapse
Affiliation(s)
- Thomas Wälchli
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland.
- Group of Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada.
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, Toronto, ON, Canada.
| | - Jeroen Bisschop
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
- Group of Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, Toronto, ON, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB & Department of Oncology, KU Leuven, Leuven, Belgium
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
- Laboratory of Angiogenesis and Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Gelareh Zadeh
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Philippe P Monnier
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Donald K. Johnson Research Institute, Krembil Research Institute, Krembil Discovery Tower, Toronto, ON, Canada
- Department of Ophthalmology and Vision Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Katrien De Bock
- Laboratory of Exercise and Health, Department of Health Science and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Ivan Radovanovic
- Group of Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, Toronto, ON, Canada
| |
Collapse
|
8
|
Wang T, Rho O, Eguiarte-Solomon F, DiGiovanni J. Twist1 as a target for prevention of cutaneous squamous cell carcinoma. Mol Carcinog 2023; 62:62-76. [PMID: 36373194 PMCID: PMC9772054 DOI: 10.1002/mc.23482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022]
Abstract
Cutaneous squamous cell carcinoma (cSCC) represents an important clinical problem requiring novel approaches for both prevention and treatment. The transcription factor, Twist-related protein 1 (Twist1), has been identified as having a key mechanistic role in the development and progression of cSCC. Studies in relevant mouse models of cSCC have shown that Twist1 regulates epithelial-mesenchymal transition (EMT) and stemness driving progression and metastasis of cSCC. In addition, further research has shown that Twist1 regulates the balance between keratinocyte proliferation and differentiation and therefore impacts earlier stages of cSCC development. Through use of keratinocyte specific Twist1 knockout models, a role for this gene in keratinocyte stem cell homeostasis has been revealed. As a transcription factor, Twist1 regulates a large number of genes both in a positive, as well as a negative manner across several interdependent pathways. Studies in keratinocyte specific knockout models have shown that Twist1 upregulates the expression of genes involved in proliferation, stemness, and EMT while downregulating the expression of genes associated with differentiation. Furthermore, a number of compounds, including naturally occurring compounds, have been identified that target Twist1 and can block its effects in cancer cells and in keratinocytes in vivo. Collectively, the current understanding of Twist1 function in cSCC development and progression suggests that it represents a potential target for prevention and treatment of cSCC.
Collapse
Affiliation(s)
- Tingzeng Wang
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78723, United States
| | - Okkyung Rho
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78723, United States
| | - Fernando Eguiarte-Solomon
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78723, United States
| | - John DiGiovanni
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78723, United States
- Center for Molecular Carcinogenesis and Toxicology, The University of Texas at Austin, Austin, TX 78723, United States
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, United States
| |
Collapse
|
9
|
Izadpanah MH, Forghanifard MM. TWIST1 Plays Role in Expression of Stemness State Markers in ESCC. Genes (Basel) 2022; 13:genes13122369. [PMID: 36553636 PMCID: PMC9777594 DOI: 10.3390/genes13122369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Stemness markers play critical roles in the maintenance of key properties of embryonic stem cells (ESCs), including the pluripotency, stemness state, and self-renewal capacities, as well as cell fate decision. Some of these features are present in cancer stem cells (CSCs). TWIST1, as a bHLH transcription factor oncogene, is involved in the epithelial-mesenchymal transition (EMT) process in both embryonic and cancer development. Our aim in this study was to investigate the functional correlation between TWIST1 and the involved genes in the process of CSCs self-renewal in human esophageal squamous cell carcinoma (ESCC) line KYSE-30. METHODS TWIST1 overexpression was enforced in the ESCC KYSE-30 cells using retroviral vector containing the specific pruf-IRES-GFP-hTWIST1 sequence. Following RNA extraction and cDNA synthesis, the mRNA expression profile of TWIST1 and the stem cell markers, including BMI1, CRIPTO1, DPPA2, KLF4, SOX2, NANOG, and MSI1, were assessed using relative comparative real-time PCR. RESULTS Ectopic expression of TWIST1 in KYSE-30 cells resulted in an increased expression of TWIST1 compared to control GFP cells by nearly 9-fold. Transduction of TWIST1-retroviral particles caused a significant enhancement in BMI1, CRIPTO1, DPPA2, KLF4, and SOX2 mRNA expression, approximately 4.5-, 3.2-, 5.5-, 3.5-, and 3.7-folds, respectively, whereas this increased TWIST1 expression caused no change in the mRNA expression of NANOG and MSI1 genes. CONCLUSIONS TWIST1 gene ectopic expression in KYSE-30 cells enhanced the level of cancer stem cell markers' mRNA expression. These results may emphasize the role of TWIST1 in the self-renewal process and may corroborate the involvement of TWIST1 in the stemness state capacity of ESCC cell line KYSE-30, as well as its potential as a therapeutic target.
Collapse
Affiliation(s)
- Mohammad Hossein Izadpanah
- Division of Human Genetics, Immunology Research Center, Avicenna Research Institute, Mashhad University of Medical Sciences, Mashhad 9196773117, Iran
| | - Mohammad Mahdi Forghanifard
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan 3671637849, Iran
- Correspondence: or ; Tel.: +98-912-711-6027
| |
Collapse
|
10
|
Xu M, Zhang T, Xia R, Wei Y, Wei X. Targeting the tumor stroma for cancer therapy. Mol Cancer 2022; 21:208. [PMID: 36324128 PMCID: PMC9628074 DOI: 10.1186/s12943-022-01670-1] [Citation(s) in RCA: 135] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/13/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
Tumors are comprised of both cancer cells and surrounding stromal components. As an essential part of the tumor microenvironment, the tumor stroma is highly dynamic, heterogeneous and commonly tumor-type specific, and it mainly includes noncellular compositions such as the extracellular matrix and the unique cancer-associated vascular system as well as a wide variety of cellular components including activated cancer-associated fibroblasts, mesenchymal stromal cells, pericytes. All these elements operate with each other in a coordinated fashion and collectively promote cancer initiation, progression, metastasis and therapeutic resistance. Over the past few decades, numerous studies have been conducted to study the interaction and crosstalk between stromal components and neoplastic cells. Meanwhile, we have also witnessed an exponential increase in the investigation and recognition of the critical roles of tumor stroma in solid tumors. A series of clinical trials targeting the tumor stroma have been launched continually. In this review, we introduce and discuss current advances in the understanding of various stromal elements and their roles in cancers. We also elaborate on potential novel approaches for tumor-stroma-based therapeutic targeting, with the aim to promote the leap from bench to bedside.
Collapse
Affiliation(s)
- Maosen Xu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Tao Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Ruolan Xia
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China.
| |
Collapse
|
11
|
Tian S, Xu X, Yang X, Fan L, Jiao Y, Zheng M, Zhang S. Roles of follistatin-like protein 3 in human non-tumor pathophysiologies and cancers. Front Cell Dev Biol 2022; 10:953551. [PMID: 36325361 PMCID: PMC9619213 DOI: 10.3389/fcell.2022.953551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 10/07/2022] [Indexed: 11/13/2022] Open
Abstract
Follistatin-like protein 3 (FSTL3) is a type of FSTLs. By interacting with a disintegrin and metalloproteinase 12 (ADAM12), transforming growth factor-β ligands (activin, myostatin and growth differentiation factor (GDF) 11), FSTL3 can either activate or inhibit these molecules in human non-tumor pathophysiologies and cancers. The FSTL3 gene was initially discovered in patients with in B-cell chronic lymphocytic leukemia, and subsequent studies have shown that the FSTL3 protein is associated with reproductive development, insulin resistance, and hematopoiesis. FSTL3 reportedly contributes to the development and progression of many cancers by promoting tumor metastasis, facilitating angiogenesis, and inducing stem cell differentiation. This review summarizes the current pathophysiological roles of FSTL3, which may be a putative prognostic biomarker for various diseases and serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Shifeng Tian
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Xiaoyi Xu
- Department of Stomatology, Tianjin Union Medical Center, Tianjin, China
| | - Xiaohui Yang
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Linlin Fan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuqi Jiao
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| |
Collapse
|
12
|
Zhang L, Zhang SS, Wang KF, Li YH, Xu HJ, Sun KX, Ma S, Leng HM, Chen SZ, Jia WJ, Zhu XJ, Li J. Overexpression of Twist1 in vascular endothelial cells promotes pathological retinal angiogenesis in mice. Zool Res 2022; 43:64-74. [PMID: 34845879 PMCID: PMC8743260 DOI: 10.24272/j.issn.2095-8137.2021.281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 11/29/2021] [Indexed: 11/07/2022] Open
Abstract
Retinal angiogenesis is a critical process for normal retinal function. However, uncontrolled angiogenesis can lead to pathological neovascularization (NV), which is closely related to most irreversible blindness-causing retinal diseases. Understanding the molecular basis behind pathological NV is important for the treatment of related diseases. Twist-related protein 1 (TWIST1) is a well-known transcription factor and principal inducer of epithelial-mesenchymal transition (EMT) in many human cancers. Our previous study showed that Twist1 expression is elevated in pathological retinal NV. To date, however, the role of TWIST1 in retinal pathological angiogenesis remains to be elucidated. To study the role of TWIST1 in pathological retinal NV and identify specific molecular targets for antagonizing pathological NV, we generated an inducible vascular endothelial cell (EC)-specific Twist1 transgenic mouse model ( Tg-Twist1 iEC+ ). Whole-mount retinas from Tg-Twist1 iEC+ mice showed retarded vascular progression and increased vascular density in the front end of the growing retinal vasculature, as well as aneurysm-like pathological retinal NV. Furthermore, overexpression of Twist1 in the ECs promoted cell proliferation but disturbed cell polarity, thus leading to uncontrolled retinal angiogenesis. TWIST1 promoted pathological NV by activating the Wnt/β-catenin signaling pathway and inducing the expression of NV formation-related genes, thereby acting as a 'valve' in the regulation of pathological angiogenesis. This study identified the critical role of TWIST1 in retinal pathological NV, thus providing a potential therapeutic target for pathological NV.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Ophthalmology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences, Chengdu, Sichuan 610072, China
- Qinghai Key Laboratory of Qinghai-Tibetan Plateau Biological Resources, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai 810008, China
- University of Chinese Academy of Science, Beijing 100049, China
| | - Shan-Shan Zhang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences, Chengdu, Sichuan 610072, China
| | - Kai-Fang Wang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences, Chengdu, Sichuan 610072, China
| | - Yi-Hui Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences, Chengdu, Sichuan 610072, China
| | - Hui-Juan Xu
- Department of Ophthalmology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences, Chengdu, Sichuan 610072, China
| | - Kuan-Xiang Sun
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences, Chengdu, Sichuan 610072, China
| | - Shi Ma
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences, Chengdu, Sichuan 610072, China
| | - Hong-Mei Leng
- Department of Ophthalmology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Si-Zhu Chen
- Department of Ophthalmology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Wen-Jing Jia
- Qinghai Key Laboratory of Qinghai-Tibetan Plateau Biological Resources, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai 810008, China
- University of Chinese Academy of Science, Beijing 100049, China
| | - Xian-Jun Zhu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences, Chengdu, Sichuan 610072, China
- Qinghai Key Laboratory of Qinghai-Tibetan Plateau Biological Resources, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai 810008, China
- University of Chinese Academy of Science, Beijing 100049, China
| | - Jie Li
- Department of Ophthalmology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences, Chengdu, Sichuan 610072, China. E-mail:
| |
Collapse
|
13
|
Hunyenyiwa T, Hendee K, Matus K, Kyi P, Mammoto T, Mammoto A. Obesity Inhibits Angiogenesis Through TWIST1-SLIT2 Signaling. Front Cell Dev Biol 2021; 9:693410. [PMID: 34660572 PMCID: PMC8511494 DOI: 10.3389/fcell.2021.693410] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 08/30/2021] [Indexed: 01/22/2023] Open
Abstract
Angiogenesis is required for functional adipose tissue maintenance, remodeling, and expansion. Physiologically balanced adipogenesis and angiogenesis are inhibited in subcutaneous adipose tissue in obese humans. However, the mechanism by which angiogenesis is inhibited in obese adipose tissue is not fully understood. Transcription factor TWIST1 controls angiogenesis and vascular function. TWIST1 expression is lower in obese human adipose tissues. Here, we have demonstrated that angiogenesis is inhibited in endothelial cells (ECs) isolated from adipose tissues of obese humans through TWIST1-SLIT2 signaling. The levels of TWIST1 and SLIT2 are lower in ECs isolated from obese human adipose tissues compared to those from lean tissues. Knockdown of TWIST1 in lean human adipose ECs decreases, while overexpression of TWIST1 in obese adipose ECs restores SLIT2 expression. DNA synthesis and cell migration are inhibited in obese adipose ECs and the effects are restored by TWIST1 overexpression. Obese adipose ECs also inhibit blood vessel formation in the gel subcutaneously implanted in mice, while these effects are restored when gels are mixed with SLIT2 or supplemented with ECs overexpressing TWIST1. These findings suggest that obesity impairs adipose tissue angiogenesis through TWIST1-SLIT2 signaling.
Collapse
Affiliation(s)
- Tendai Hunyenyiwa
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Kathryn Hendee
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Kienna Matus
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Priscilla Kyi
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Tadanori Mammoto
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Akiko Mammoto
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
14
|
Sphyris N, King C, Hoar J, Werden SJ, Vijay GV, Miura N, Gaharwar A, Sarkar TR. Carcinoma cells that have undergone an epithelial-mesenchymal transition differentiate into endothelial cells and contribute to tumor growth. Oncotarget 2021; 12:823-844. [PMID: 33889304 PMCID: PMC8057273 DOI: 10.18632/oncotarget.27940] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/25/2021] [Indexed: 12/23/2022] Open
Abstract
Hypoxia stimulates neoangiogenesis, promoting tumor outgrowth, and triggers the epithelial-mesenchymal transition (EMT), which bestows cells with mesenchymal traits and multi-lineage differentiation potential. Here, we investigated whether EMT can confer endothelial attributes upon carcinoma cells, augmenting tumor growth and vascularization. Following orthotopic implantation of MCF-7 human epithelial breast cancer cells into mice, tumors of different sizes were immunostained for markers of hypoxia and EMT. Larger tumors were well-vascularized with CD31-positive cells of human origin. Hypoxic regions, demarcated by HIF-1α staining, exhibited focal areas of E-cadherin loss and elevated levels of vimentin and the EMT-mediator FOXC2. Implantation of MCF-7 cells, co-mixed with human mammary epithelial (HMLE) cells overexpressing the EMT-inducer Snail, markedly potentiated tumor growth and vascularization, compared with MCF-7 cells injected alone or co-mixed with HMLE-vector cells. Intra-tumoral vessels contained CD31-positive cells derived from either donor cell type. FOXC2 knockdown abrogated the potentiating effects of HMLE-Snail cells on MCF-7 tumor growth and vascularization, and compromised endothelial transdifferentiation of mesenchymal cells cultured in endothelial growth medium. Hence, cells that have undergone EMT can promote tumor growth and neovascularization either indirectly, by promoting endothelial transdifferentiation of carcinoma cells, or directly, by acquiring an endothelial phenotype, with FOXC2 playing key roles in these processes.
Collapse
Affiliation(s)
- Nathalie Sphyris
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Present address: Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK.,These authors contributed equally to this work
| | - Cody King
- Department of Biochemistry, Texas A&M University, College Station, TX, USA
| | - Jonathan Hoar
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Steven J Werden
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Geraldine V Vijay
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naoyuki Miura
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Akhilesh Gaharwar
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Tapasree Roy Sarkar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Biology, Texas A&M University, College Station, TX, USA.,These authors contributed equally to this work
| |
Collapse
|
15
|
Twist1 signaling in age-dependent decline in angiogenesis and lung regeneration. Aging (Albany NY) 2021; 13:7781-7799. [PMID: 33764901 PMCID: PMC8034921 DOI: 10.18632/aging.202875] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 03/14/2021] [Indexed: 12/11/2022]
Abstract
Angiogenesis – the formation of new blood capillaries- is impaired in aging animals and contributes to the pathogenesis of age-related diseases. A transcription factor, Twist1, contributes to the pathogenesis of age- and angiogenesis-related diseases such as pulmonary fibrosis and atherosclerosis. However, the mechanism by which Twist1 controls age-dependent decline in angiogenesis remains unclear. In this report, we have demonstrated that the levels of Twist1 are higher, while the expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) that stimulates angiogenesis, is lower in endothelial cells (ECs) isolated from aged human adipose tissues and mouse lungs compared to those from young tissues. Knockdown of Twist1 in aged human ECs increases the levels of PGC1α and angiogenic factor receptor, vascular endothelial growth factor receptor (VEGFR2), and restores EC proliferation and migration, while inhibition of PGC1α suppresses these effects. Knockdown of Twist1 in supplemented aged ECs also restores vascular networks in the subcutaneously implanted gel, while these effects are abrogated by knockdown of PGC1α. Age-dependent inhibition of post-pneumonectomy (PNX) lung growth is suppressed in Tie2-specific Twist1 conditional knockout mouse lungs, in which VEGFR2 expression increases after PNX. These results suggest that upregulation of endothelial Twist1 mediates age-dependent decline in angiogenesis and regenerative lung growth.
Collapse
|
16
|
Fattahi F, Saeednejad Zanjani L, Vafaei S, Habibi Shams Z, Kiani J, Naseri M, Gheytanchi E, Madjd Z. Expressions of TWIST1 and CD105 markers in colorectal cancer patients and their association with metastatic potential and prognosis. Diagn Pathol 2021; 16:26. [PMID: 33752711 PMCID: PMC7983220 DOI: 10.1186/s13000-021-01088-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 03/10/2021] [Indexed: 01/06/2023] Open
Abstract
Background TWIST1 and CD105, which contribute to tumor malignancy, are overexpressed in cancers. Accordingly, TWIST1 enhances epithelial-to-mesenchymal transition (EMT) and promotes the formation of cancer stem cells (CSCs). Also, CD105 is a neoangiogenesis marker in endothelial cells, which is introduced as a CSC marker in tumoral epithelial cells in several types of cancers. The present study was aimed to investigate expressions of TWIST1 and CD105 in colorectal cancer (CRC) patients. Methods Expressions of TWIST1 and CD105 in 250 CRC tissue samples were evaluated using immunohistochemistry on tissue microarrays (TMAs). In this regard, TWIST1 expression was investigated in the subcellular locations (cytoplasm and nucleus), while CD105 was mapped in endothelial cells and cytoplasmic tumor cells of CRC tissues. The association between the expression of these markers and clinicopathological parameters, as well as survival outcomes were analyzed. Results Results indicate a statistically significant association between higher nuclear expression levels of TWIST1 and distant metastases in CRC (P = 0.040) patients. In addition, it was shown that the increased nuclear expression of TWIST1 had a poor prognostic value for disease-specific survival (DSS) and progression-free survival (PFS) (P = 0.042, P = 0.043, respectively) in patients with CRC. Moreover, analysis of CD105 expression level has revealed that there is a statistically significant association between the increased expression of CD105 in tumoral epithelial cells and more advanced TNM stage (P = 0.050). Conclusions Our results demonstrate that nuclear TWIST1 and cytoplasmic CD105 expressions in tumor cells had associations with more aggressive tumor behavior and more advanced diseases in CRC cases.
Collapse
Affiliation(s)
- Fahimeh Fattahi
- Oncopathology Research Center, Iran University of Medical Sciences , Postal Code: 14496-14530, Hemmat Street (Highway), Next to Milad Tower, Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Leili Saeednejad Zanjani
- Oncopathology Research Center, Iran University of Medical Sciences , Postal Code: 14496-14530, Hemmat Street (Highway), Next to Milad Tower, Tehran, Iran
| | - Somayeh Vafaei
- Oncopathology Research Center, Iran University of Medical Sciences , Postal Code: 14496-14530, Hemmat Street (Highway), Next to Milad Tower, Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences , Postal Code: 14496-14530, Hemmat Street (Highway), Next to Milad Tower, Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Marzieh Naseri
- Oncopathology Research Center, Iran University of Medical Sciences , Postal Code: 14496-14530, Hemmat Street (Highway), Next to Milad Tower, Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elmira Gheytanchi
- Oncopathology Research Center, Iran University of Medical Sciences , Postal Code: 14496-14530, Hemmat Street (Highway), Next to Milad Tower, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences , Postal Code: 14496-14530, Hemmat Street (Highway), Next to Milad Tower, Tehran, Iran. .,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran. .,Department of Pathology, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Unlocking the Secrets of Cancer Stem Cells with γ-Secretase Inhibitors: A Novel Anticancer Strategy. Molecules 2021; 26:molecules26040972. [PMID: 33673088 PMCID: PMC7917912 DOI: 10.3390/molecules26040972] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 12/26/2022] Open
Abstract
The dysregulation of Notch signaling is associated with a wide variety of different human cancers. Notch signaling activation mostly relies on the activity of the γ-secretase enzyme that cleaves the Notch receptors and releases the active intracellular domain. It is well-documented that γ-secretase inhibitors (GSIs) block the Notch activity, mainly by inhibiting the oncogenic activity of this pathway. To date, several GSIs have been introduced clinically for the treatment of various diseases, such as Alzheimer's disease and various cancers, and their impacts on Notch inhibition have been found to be promising. Therefore, GSIs are of great interest for cancer therapy. The objective of this review is to provide a systematic review of in vitro and in vivo studies for investigating the effect of GSIs on various cancer stem cells (CSCs), mainly by modulation of the Notch signaling pathway. Various scholarly electronic databases were searched and relevant studies published in the English language were collected up to February 2020. Herein, we conclude that GSIs can be potential candidates for CSC-targeting therapy. The outcome of our study also indicates that GSIs in combination with anticancer drugs have a greater inhibitory effect on CSCs.
Collapse
|
18
|
Fasoulakis Z, Daskalakis G, Theodora M, Antsaklis P, Sindos M, Diakosavvas M, Angelou K, Loutradis D, Kontomanolis EN. The Relevance of Notch Signaling in Cancer Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1287:169-181. [PMID: 33034032 DOI: 10.1007/978-3-030-55031-8_11] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The Notch signaling pathway controls normal embryonic development and tissue homeostasis of many cell types. It regulates cell proliferation, fate, differentiation, and cell death by short-range signaling between nearby cells that come in contact. The Notch pathway has also been critically involved in the pathobiology of a variety of malignancies, regulating cancer initiation and development, as well as early stages of cancer progression, by adjusting conserved cellular programs. Fibroblasts, an essential for tumor growth component of stroma, have also been affected by Notch regulation. Sequencing Notch gene mutations have been identified in a number of human tumors, revealing information on the progression of specific cancer types, such as ovarian cancer and melanoma, immune-associated tumors such as myeloid neoplasms, but especially in lymphocytic leukemia. Activation of the Notch can be either oncogenic or it may contain growth-suppressive functions, acting as a tumor suppressor in other hematopoietic cells, hepatocytes, skin, and pancreatic epithelium.
Collapse
Affiliation(s)
- Zacharias Fasoulakis
- National and Kapodistrian University of Athens - 1st Department of Obstetrics and Gynecology, Athens, Greece.
| | - George Daskalakis
- National and Kapodistrian University of Athens - 1st Department of Obstetrics and Gynecology, Athens, Greece
| | - Marianna Theodora
- National and Kapodistrian University of Athens - 1st Department of Obstetrics and Gynecology, Athens, Greece
| | - Panos Antsaklis
- National and Kapodistrian University of Athens - 1st Department of Obstetrics and Gynecology, Athens, Greece
| | - Michael Sindos
- National and Kapodistrian University of Athens - 1st Department of Obstetrics and Gynecology, Athens, Greece
| | - Michail Diakosavvas
- National and Kapodistrian University of Athens - 1st Department of Obstetrics and Gynecology, Athens, Greece
| | - Kyveli Angelou
- National and Kapodistrian University of Athens - 1st Department of Obstetrics and Gynecology, Athens, Greece
| | - Dimitrios Loutradis
- National and Kapodistrian University of Athens - 1st Department of Obstetrics and Gynecology, Athens, Greece
| | - Emmanuel N Kontomanolis
- Democritus University of Thrace - Department of Obstetrics and Gynecology, Alexandroupolis, Greece
| |
Collapse
|
19
|
Teixeira FCOB, Vijaya Kumar A, Kumar Katakam S, Cocola C, Pelucchi P, Graf M, Kiesel L, Reinbold R, Pavão MSG, Greve B, Götte M. The Heparan Sulfate Sulfotransferases HS2ST1 and HS3ST2 Are Novel Regulators of Breast Cancer Stem-Cell Properties. Front Cell Dev Biol 2020; 8:559554. [PMID: 33102470 PMCID: PMC7546021 DOI: 10.3389/fcell.2020.559554] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/02/2020] [Indexed: 12/18/2022] Open
Abstract
Heparan sulfate (HS) is a glycosaminoglycan found mainly in its protein-conjugated form at the cell surface and the extracellular matrix. Its high sulfation degree mediates functional interactions with positively charged amino acids in proteins. 2-O sulfation of iduronic acid and 3-O sulfation of glucosamine in HS are mediated by the sulfotransferases HS2ST and HS3ST, respectively, which are dysregulated in several cancers. Both sulfotransferases regulate breast cancer cell viability and invasion, but their role in cancer stem cells (CSCs) is unknown. Breast CSCs express characteristic markers such as CD44+/CD24−/low, CD133 and ALDH1 and are involved in tumor initiation, formation, and recurrence. We studied the influence of HS2ST1 and HS3ST2 overexpression on the CSC phenotype in breast cancer cell lines representative of the triple-negative (MDA-MB-231) and hormone-receptor positive subtype (MCF-7). The CD44+/CD24−/low phenotype was significantly reduced in MDA-MB-231 cells after overexpression of both enzymes, remaining unaltered in MCF-7 cells. ALDH1 activity was increased after HS2ST1 and HS3ST2 overexpression in MDA-MB-231 cells and reduced after HS2ST1 overexpression in MCF-7 cells. Colony and spheroid formation were increased after HS2ST1 and HS3ST2 overexpression in MCF-7 cells. Moreover, MDA-MB-231 cells overexpressing HS2ST1 formed more colonies and could not generate spheres. The phenotypic changes were associated with complex changes in the expression of the stemness-associated notch and Wnt-signaling pathways constituents, syndecans, heparanase and Sulf1. The results improve our understanding of breast CSC function and mark a subtype-specific impact of HS modifications on the CSC phenotype of triple-negative and hormone receptor positive breast cancer model cell lines.
Collapse
Affiliation(s)
- Felipe C O B Teixeira
- Instituto de Bioquímica Médica Leopoldo de Meis, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Archana Vijaya Kumar
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Sampath Kumar Katakam
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Cinzia Cocola
- Istituto di Tecnologie Biomediche, Consiglio Nazionale delle Ricerche, Segrate, Italy
| | - Paride Pelucchi
- Istituto di Tecnologie Biomediche, Consiglio Nazionale delle Ricerche, Segrate, Italy
| | - Monika Graf
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Rolland Reinbold
- Istituto di Tecnologie Biomediche, Consiglio Nazionale delle Ricerche, Segrate, Italy
| | - Mauro S G Pavão
- Instituto de Bioquímica Médica Leopoldo de Meis, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Burkhard Greve
- Department of Radiotherapy and Radiooncology, University Hospital of Münster, Münster, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| |
Collapse
|
20
|
Kuang XY, Ren Y, Chen C, Su J, Li HM, Liu SJ, Sun TJ, Mu DY, Lu J, Chen L, Qu HD, Cui YH, Yu AY, Yao XH. Quantitative analysis for the differences in vasculogenic activity and sensitivity to angiogenic stimulants between human glioma cells and normal endothelial cells. Brain Res 2020; 1748:147082. [PMID: 32866544 DOI: 10.1016/j.brainres.2020.147082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/19/2020] [Accepted: 08/24/2020] [Indexed: 01/10/2023]
Abstract
Neovascularization is a histological feature of glioma, especially of glioblastoma (GBM), being associated with tumor invasiveness and poor prognosis. However, current anti-angiogenic therapies targeting vascular endothelial cells (ECs), has exhibited poor efficacy in some GBM cases. This may be at least partially attributed to the potential of glioblastoma cells to construct blood supply chain via vasculogenic mimicry or endothelial differentiation. This study aims to explore differences in vasculogenic activity and sensitivity to angiogenic stimulants between normal human ECs and glioma cells of different grades. We found that grade IV U87 GBM cells showed highly inducible vasculogenic activity either in the orthotopic xenograft model or under in vitro angiogenic stimulants as compared with grade II CHG5 glioma cells. The hypoxia mimetic more strongly induced in vitro vasculogenic capacity and endothelial marker expression of U87 GBM cells than the stimulation with multiple proangiogenic growth factors (vascular endothelial growth factor, basic fibroblast growth factor and epidermal growth factor). In contrast, proangiogenic effect of hypoxia on human umbilical vein endothelial cells (HUVECs) was weaker than on U87 GBM cells. In addition, it was also observed that the in vitro vasculogenic process of U87 cells started later but lasted longer than that of HUVECs. These results demonstrate that when compared with normal ECs, high-grade glioma cells basically possess weaker vasculogenic activity, but exhibit higher sensitivity and longer-lasting response to angiogenic stimulants, especially to hypoxia. This may be helpful to develop novel anti-angiogenic strategies targeting both vascular ECs and vasculogenic glioma cells.
Collapse
Affiliation(s)
- Xiao-Yan Kuang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China; Experiment Center for Science and Technology, Zunyi Medical and Pharmaceutical College, Zunyi 563006, China
| | - Yong Ren
- Department of Pathology, Central Theater Command General Hospital, Wuhan 430070, China
| | - Cong Chen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jun Su
- Department of Pathology, the First Affiliated Hospital of Zunyi Medical University, Zunyi 563099, China
| | - Hua-Mei Li
- Experiment Center for Science and Technology, Zunyi Medical and Pharmaceutical College, Zunyi 563006, China
| | - Si-Jia Liu
- Emergency Department, The First Affiliated Hospital of Zunyi Medical University, Zunyi 563099, China
| | - Tian-Jing Sun
- Emergency Department, The First Affiliated Hospital of Zunyi Medical University, Zunyi 563099, China
| | - Dong-Yun Mu
- Experiment Center for Science and Technology, Zunyi Medical and Pharmaceutical College, Zunyi 563006, China
| | - Jing Lu
- Experiment Center for Science and Technology, Zunyi Medical and Pharmaceutical College, Zunyi 563006, China
| | - Lan Chen
- Experiment Center for Science and Technology, Zunyi Medical and Pharmaceutical College, Zunyi 563006, China
| | - Hang-Da Qu
- Experiment Center for Science and Technology, Zunyi Medical and Pharmaceutical College, Zunyi 563006, China
| | - You-Hong Cui
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - An-Yong Yu
- Emergency Department, The First Affiliated Hospital of Zunyi Medical University, Zunyi 563099, China.
| | - Xiao-Hong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
21
|
Snail promotes the generation of vascular endothelium by breast cancer cells. Cell Death Dis 2020; 11:457. [PMID: 32541667 PMCID: PMC7295784 DOI: 10.1038/s41419-020-2651-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 12/13/2022]
Abstract
A further understanding of tumor angiogenesis is urgently needed due to the limited therapeutic efficacy of anti-angiogenesis agents. However, the origin of endothelial cells (EC) in tumors remains widely elusive and controversial. Snail has been thoroughly elucidated as a master regulator of the epithelial-mesenchymal transition (EMT), but its role in endothelium generation is not yet established. In this study, we reported a new and unexpected function of Snail in endothelium generation by breast cancer cells. We showed that high Snail-expressing breast cancer cells isolated from patients showed more endothelium generated from these cells. Expression of Snail was positively correlated with endothelial markers in breast cancer patients. The ectopic expression of Snail induced endothelial marker expression, tube formation and DiI-AcLDL uptake of breast cancer cells in vitro, and enhanced tumor growth and microvessel density in vivo. Snail-mediated endothelium generation depended on VEGF and Sox2. Mechanistically, Snail promoted the expression of VEGF and Sox2 through recruiting the p300 activator complex to these promoters. We showed the dual function of Snail in tumor initiation and angiogenesis in vivo and in vitro through activation of Sox2 and VEGF, suggesting Snail may be an ideal target for cancer therapy.
Collapse
|
22
|
Kaushik K, Das A. TWIST1-Reprogrammed Endothelial Cell Transplantation Potentiates Neovascularization-Mediated Diabetic Wound Tissue Regeneration. Diabetes 2020; 69:1232-1247. [PMID: 32234721 DOI: 10.2337/db20-0138] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 03/23/2020] [Indexed: 11/13/2022]
Abstract
Hypovascularized diabetic nonhealing wounds are due to reduced number and impaired physiology of endogenous endothelial progenitor cell (EPC) population that limits their recruitment and mobilization at the wound site. For enrichment of the EPC repertoire from nonendothelial precursors, abundantly available mesenchymal stromal cells (MSC) were reprogrammed into induced endothelial cells (iEC). We identified cell signaling molecular targets by meta-analysis of microarray data sets. BMP-2 induction leads to the expression of inhibitory Smad 6/7-dependent negative transcriptional regulation of ID1, rendering the latter's reduced binding to TWIST1 during transdifferentiation of Wharton jelly-derived MSC (WJ-MSC) into iEC. TWIST1, in turn, regulates endothelial gene transcription, positively of proangiogenic KDR and negatively, in part, of antiangiogenic SFRP4 Twist1 reprogramming enhanced the endothelial lineage commitment of WJ-MSC and increased the vasculogenic potential of reprogrammed endothelial cells (rEC). Transplantation of stable TWIST1 rEC into a type 1 and 2 diabetic full-thickness splinted wound healing murine model enhanced the microcirculatory blood flow and accelerated the wound tissue regeneration. An increased or decreased colocalization of GFP with KDR/SFRP4 and CD31 in the regenerated diabetic wound bed with TWIST1 overexpression or silencing (piLenti-TWIST1-shRNA-GFP), respectively, further confirmed improved neovascularization. This study depicted the reprogramming of WJ-MSC into rEC using unique transcription factor TWIST1 for an efficacious cell transplantation therapy to induce neovascularization-mediated diabetic wound tissue regeneration.
Collapse
Affiliation(s)
- Komal Kaushik
- Department of Applied Biology, Council of Scientific and Industrial Research-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific and Innovative Research, Council of Scientific and Industrial Research-Indian Institute of Chemical Technology Campus, Hyderabad, India
| | - Amitava Das
- Department of Applied Biology, Council of Scientific and Industrial Research-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific and Innovative Research, Council of Scientific and Industrial Research-Indian Institute of Chemical Technology Campus, Hyderabad, India
| |
Collapse
|
23
|
Mammoto A, Hendee K, Muyleart M, Mammoto T. Endothelial Twist1-PDGFB signaling mediates hypoxia-induced proliferation and migration of αSMA-positive cells. Sci Rep 2020; 10:7563. [PMID: 32371931 PMCID: PMC7200682 DOI: 10.1038/s41598-020-64298-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 04/10/2020] [Indexed: 02/01/2023] Open
Abstract
Remodeling of distal pulmonary arterioles (PAs) associated with marked accumulation of pulmonary artery smooth muscle cells (PASMCs) represents one of the major pathologic features of pulmonary hypertension (PH). We have reported that the transcription factor Twist1 mediates hypoxia-induced PH. However, the mechanism by which endothelial Twist1 stimulates SMC accumulation to distal PAs in PH remains unclear. Here, we have demonstrated that Twist1 overexpression increases the expression of platelet-derived growth factor (PDGFB) in human pulmonary arterial endothelial (HPAE) cells. Hypoxia upregulates the levels of Twist1 and PDGFB in HPAE cells. When we implant hydrogel supplemented with endothelial cells (ECs) on the mouse lung, these ECs form vascular lumen structures and hypoxia upregulates PDGFB expression and stimulates accumulation of αSMA–positive cells in the gel, while knockdown of endothelial Twist1 suppresses the effects. The levels of Twist1 and PDGFB are higher in PAE cells isolated from idiopathic pulmonary arterial hypertension (IPAH) patients compared to those from healthy controls. IPAH patient-derived PAE cells stimulate accumulation of αSMA–positive cells in the implanted gel, while Twist1 knockdown in PAE cells inhibits the effects. Endothelial Twist1-PDGFB signaling plays a key role in αSMA–positive cell proliferation and migration in PH.
Collapse
Affiliation(s)
- Akiko Mammoto
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, 53226, United States. .,Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, United States.
| | - Kathryn Hendee
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, 53226, United States
| | - Megan Muyleart
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, 53226, United States
| | - Tadanori Mammoto
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, 53226, United States.
| |
Collapse
|
24
|
Haibe Y, Kreidieh M, El Hajj H, Khalifeh I, Mukherji D, Temraz S, Shamseddine A. Resistance Mechanisms to Anti-angiogenic Therapies in Cancer. Front Oncol 2020; 10:221. [PMID: 32175278 PMCID: PMC7056882 DOI: 10.3389/fonc.2020.00221] [Citation(s) in RCA: 242] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
Tumor growth and metastasis rely on tumor vascular network for the adequate supply of oxygen and nutrients. Tumor angiogenesis relies on a highly complex program of growth factor signaling, endothelial cell (EC) proliferation, extracellular matrix (ECM) remodeling, and stromal cell interactions. Numerous pro-angiogenic drivers have been identified, the most important of which is the vascular endothelial growth factor (VEGF). The importance of pro-angiogenic inducers in tumor growth, invasion and extravasation make them an excellent therapeutic target in several types of cancers. Hence, the number of anti-angiogenic agents developed for cancer treatment has risen over the past decade, with at least eighty drugs being investigated in preclinical studies and phase I-III clinical trials. To date, the most common approaches to the inhibition of the VEGF axis include the blockade of VEGF receptors (VEGFRs) or ligands by neutralizing antibodies, as well as the inhibition of receptor tyrosine kinase (RTK) enzymes. Despite promising preclinical results, anti-angiogenic monotherapies led only to mild clinical benefits. The minimal benefits could be secondary to primary or acquired resistance, through the activation of alternative mechanisms that sustain tumor vascularization and growth. Mechanisms of resistance are categorized into VEGF-dependent alterations, non-VEGF pathways and stromal cell interactions. Thus, complementary approaches such as the combination of these inhibitors with agents targeting alternative mechanisms of blood vessel formation are urgently needed. This review provides an updated overview on the pathophysiology of angiogenesis during tumor growth. It also sheds light on the different pro-angiogenic and anti-angiogenic agents that have been developed to date. Finally, it highlights the preclinical evidence for mechanisms of angiogenic resistance and suggests novel therapeutic approaches that might be exploited with the ultimate aim of overcoming resistance and improving clinical outcomes for patients with cancer.
Collapse
Affiliation(s)
- Yolla Haibe
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
| | - Malek Kreidieh
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
| | - Hiba El Hajj
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut-Medical Center, Beirut, Lebanon
| | - Ibrahim Khalifeh
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Deborah Mukherji
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
| | - Sally Temraz
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
| | - Ali Shamseddine
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
| |
Collapse
|
25
|
Mahmoud M, Souilhol C, Serbanovic-Canic J, Evans P. GATA4-Twist1 Signalling in Disturbed Flow-Induced Atherosclerosis. Cardiovasc Drugs Ther 2020; 33:231-237. [PMID: 30809744 DOI: 10.1007/s10557-019-06863-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Endothelial cell (EC) dysfunction (enhanced inflammation, proliferation and permeability) is the initial trigger for atherosclerosis. Atherosclerosis shows preferential development near branches and bends exposed to disturbed blood flow. By contrast, sites that are exposed to non-disturbed blood flow are atheroprotected. Disturbed flow promotes atherosclerosis by promoting EC dysfunction. Blood flow controls EC function through transcriptional and post-transcriptional mechanisms that are incompletely understood. METHODS AND RESULTS We identified the developmental transcription factors Twist1 and GATA4 as being enriched in EC at disturbed flow, atheroprone regions of the porcine aorta in a microarray study. Further work using the porcine and murine aortae demonstrated that Twist1 and GATA4 expression was enhanced at the atheroprone, disturbed flow sites in vivo. Using controlled in vitro flow systems, the expression of Twist1 and GATA4 was enhanced under disturbed compared to non-disturbed flow in cultured cells. Disturbed flow promoted Twist1 expression through a GATA4-mediated transcriptional mechanism as revealed by a series of in vivo and in vitro studies. GATA4-Twist1 signalling promoted EC proliferation, inflammation, permeability and endothelial-to-mesenchymal transition (EndoMT) under disturbed flow, leading to atherosclerosis development, as shown in a combination of in vitro and in vivo studies using GATA4 and Twist1-specific siRNA and EC-specific GATA4 and Twist1 Knock out (KO) mice. CONCLUSIONS We revealed that GATA4-Twist1-Snail signalling triggers EC dysfunction and atherosclerosis; this work could lead to the development of novel anti-atherosclerosis therapeutics.
Collapse
Affiliation(s)
- Marwa Mahmoud
- Department of Biomedical Engineering, The City College of New York, New York, NY, 10031, USA.
| | - Celine Souilhol
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Jovana Serbanovic-Canic
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Paul Evans
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| |
Collapse
|
26
|
Sobierajska K, Ciszewski WM, Sacewicz-Hofman I, Niewiarowska J. Endothelial Cells in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1234:71-86. [PMID: 32040856 DOI: 10.1007/978-3-030-37184-5_6] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Angiogenesis is a critical process required for tumor progression. Newly formed blood vessels provide nutrition and oxygen to the tumor contributing to its growth and development. However, endothelium also plays other functions that promote tumor metastasis. It is involved in intravasation, which allows invasive cancer cells to translocate into the blood vessel lumen. This phenomenon is an important stage for cancer metastasis. Besides direct association with cancer development, endothelial cells are one of the main sources of cancer-associated fibroblasts (CAFs). The heterogeneous group of CAFs is the main inductor of migration and invasion abilities of cancer cells. Therefore, the endothelium is also indirectly responsible for metastasis. Considering the above, the endothelium is one of the important targets of anticancer therapy. In the chapter, we will present mechanisms regulating endothelial function, dependent on cancer and cancer niche cells. We will focus on possibilities of suppressing pro-metastatic endothelial functions, applied in anti-cancer therapies.
Collapse
Affiliation(s)
| | | | | | - Jolanta Niewiarowska
- Department of Molecular Cell Mechanisms, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
27
|
Shaping of the Tumor Microenvironment by Notch Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1223:1-16. [PMID: 32030682 DOI: 10.1007/978-3-030-35582-1_1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The tumor microenvironment (TME) has become a major concern of cancer research both from a basic and a therapeutic point of view. Understanding the effect of a signaling pathway-and thus the effect of its targeting-in every aspect of the microenvironment is a prerequisite to predict and analyze the effect of a therapy. The Notch signaling pathway is involved in every component of the TME as well as in the interaction between the different parts of the TME. This review aims at describing how Notch signaling is impacting the TME and the consequences this may have when modulating Notch signaling in a therapeutic perspective.
Collapse
|
28
|
Wang XX, Yin GQ, Zhang ZH, Rong ZH, Wang ZY, Du DD, Wang YD, Gao RX, Xian GZ. TWIST1 transcriptionally regulates glycolytic genes to promote the Warburg metabolism in pancreatic cancer. Exp Cell Res 2020; 386:111713. [PMID: 31705846 DOI: 10.1016/j.yexcr.2019.111713] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 11/04/2019] [Accepted: 11/05/2019] [Indexed: 12/28/2022]
Abstract
Reprogrammed glucose metabolism is essential for tumor initiation and development, especially for pancreatic ductal adenocarcinoma (PDAC). Most cancer cells rely on aerobic glycolysis, a phenomenon termed "the Warburg effect", to support uncontrolled proliferation and evade apoptosis. However, the direct regulators of the Warburg effect remain areas of active investigation. In this study, we found that the highly conserved transcription factor, TWIST1, is a crucial regulator of aerobic glycolysis in PDAC. Genetic silencing of TWIST1 significantly inhibited the glycolytic phenotypes of PDAC cells as revealed by reduced glucose uptake, lactate production, and extracellular acidification rate, which can be restored by re-expression of siRNA-resistant TWIST1. Moreover, tamoxifen-inducible expression of TWIST1 promoted the Warburg metabolism of PDAC cells. Mechanistically, by luciferase reporter assay and chromatin immunoprecipitation experiment, we showed that TWIST1 can directly increase the expression of several glycolytic genes, including SLC2A1, HK2, ENO1, and PKM2. Of note, the transcriptional regulation by TWIST1 was not dependent on HIF1α or c-Myc. In The Cancer Genome Atlas and Gene Expression Omnibus accession GSE15471, we confirmed that TWIST1 was closely associated with the glycolysis pathway. Collectively, our findings indicate that TWIST1 is likely to act as important regulator of the Warburg effect in PDAC.
Collapse
Affiliation(s)
- Xin-Xing Wang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong Province, China
| | - Guo-Qing Yin
- Department of General Surgery, Qingzhou People's Hospital, Qingzhou, 262500, Shandong Province, China
| | - Zhen-Hai Zhang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong Province, China
| | - Zhong-Hou Rong
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong Province, China
| | - Zhi-Yi Wang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong Province, China
| | - Dong-Dong Du
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong Province, China
| | - Ya-Dong Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Shangdong First Medical University, Jinan, 250014, Shandong Province, China
| | - Ru-Xin Gao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Shangdong First Medical University, Jinan, 250014, Shandong Province, China
| | - Guo-Zhe Xian
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong Province, China.
| |
Collapse
|
29
|
Souilhol C, Serbanovic-Canic J, Fragiadaki M, Chico TJ, Ridger V, Roddie H, Evans PC. Endothelial responses to shear stress in atherosclerosis: a novel role for developmental genes. Nat Rev Cardiol 2020; 17:52-63. [PMID: 31366922 DOI: 10.1038/s41569-019-0239-5] [Citation(s) in RCA: 290] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/04/2019] [Indexed: 01/04/2023]
Abstract
Flowing blood generates a frictional force called shear stress that has major effects on vascular function. Branches and bends of arteries are exposed to complex blood flow patterns that exert low or low oscillatory shear stress, a mechanical environment that promotes vascular dysfunction and atherosclerosis. Conversely, physiologically high shear stress is protective. Endothelial cells are critical sensors of shear stress but the mechanisms by which they decode complex shear stress environments to regulate physiological and pathophysiological responses remain incompletely understood. Several laboratories have advanced this field by integrating specialized shear-stress models with systems biology approaches, including transcriptome, methylome and proteome profiling and functional screening platforms, for unbiased identification of novel mechanosensitive signalling pathways in arteries. In this Review, we describe these studies, which reveal that shear stress regulates diverse processes and demonstrate that multiple pathways classically known to be involved in embryonic development, such as BMP-TGFβ, WNT, Notch, HIF1α, TWIST1 and HOX family genes, are regulated by shear stress in arteries in adults. We propose that mechanical activation of these pathways evolved to orchestrate vascular development but also drives atherosclerosis in low shear stress regions of adult arteries.
Collapse
Affiliation(s)
- Celine Souilhol
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Jovana Serbanovic-Canic
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Maria Fragiadaki
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Timothy J Chico
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Bateson Centre for Lifecourse Biology, University of Sheffield, Sheffield, UK
| | - Victoria Ridger
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Hannah Roddie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Paul C Evans
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK.
- Bateson Centre for Lifecourse Biology, University of Sheffield, Sheffield, UK.
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK.
| |
Collapse
|
30
|
Liu X, Li C, Yang Y, Liu X, Li R, Zhang M, Yin Y, Qu Y. Synaptotagmin 7 in twist-related protein 1-mediated epithelial - Mesenchymal transition of non-small cell lung cancer. EBioMedicine 2019; 46:42-53. [PMID: 31395502 PMCID: PMC6711891 DOI: 10.1016/j.ebiom.2019.07.071] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/28/2019] [Accepted: 07/30/2019] [Indexed: 12/18/2022] Open
Abstract
Background Twist-related protein 1 (TWIST1) plays an essential role in the carcinogenesis and metastasis of NSCLC. Our aims were to identify the molecule at the downstream of TWIST1 and to evaluate its potential as a diagnostic and a prognostic marker in NSCLC. Methods The functional genes at the downstream of TWIST1 were obtained via microarray gene expression analyses in the NSCLC cell line. The expression levels of synaptotagmin 7 (SYT7) in a cohort of patients with NSCLC (n = 154) were examined using immunohistochemistry staining and assessed by Kaplan-Meier survival analysis and Cox regression analysis. The effects of SYT7 on the tumorigenesis and metastasis of NSCLC were measured in NSCLC cells. In vivo xenograft lung cancer models were used to study the tumorigenesis role of SYT7. Findings We discovered that SYT7 is significantly altered by TWIST1 expression. We further confirmed that SYT7 protein was significantly higher in NSCLC than that in the adjacent normal lung tissue, and higher SYT7 expression was associated with poor survival of NSCLC patients. The protein level of SYT7 was positively correlated with TWIST1 in NSCLC tissue. Functional experiments indicated that SYT7 promoted proliferation, invasion, and metastasis and inhibited cell apoptosis of NSCLC cells in vitro. In vivo experiments showed that shSYT7 inhibited the xenograft tumor growth of NSCLC cells. Knocking down of SYT7 increased the expression of E-cadherin and decreased the level of N-cadherin and Vimentin in cultured cells. Interpretation Our data indicate that SYT7 is an important promoter for EMT and tumor progression in NSCLC. Fund This project was supported by grants from the Major Scientific and Technological Innovation Project of Shandong Province (2018CXGC1212), Science and Technology Foundation of Shandong Province (2014GSF118084, 2016GSF121043), Medical and Health Technology Innovation Plan of Jinan City (201805002) and the National Natural Science Foundation of China (81372333).
Collapse
Affiliation(s)
- Xiao Liu
- Department of Respiratory and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Chunyu Li
- Department of Respiratory Medicine, First Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Yie Yang
- Department of Clinical Laboratory, Qianfoshan Hospital of Shandong Province, Jinan 250012, China
| | - Xiaoxia Liu
- Department of Respiratory and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Rui Li
- Department of Respiratory and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Mengyu Zhang
- Department of Respiratory and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Yunhong Yin
- Department of Respiratory and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Yiqing Qu
- Department of Respiratory and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan 250012, China.
| |
Collapse
|
31
|
Liu Y, Ciotti GE, Eisinger-Mathason TSK. Hypoxia and the Tumor Secretome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1136:57-69. [PMID: 31201716 DOI: 10.1007/978-3-030-12734-3_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Metastasis remains the leading cause of cancer-related deaths. To date, there are no specific treatments targeting disseminated disease. New therapeutic options will become available only if we enhance our understanding of mechanisms underlying metastatic spread. A large body of literature shows that the metastatic potential of tumor cells is strongly influenced by microenvironmental cues such as low oxygen (hypoxia). Clinically, hypoxia is a hallmark of most solid tumors and is associated with increased metastasis and poor survival in a variety of cancer types. Mechanistically, hypoxia influences multiple steps within the metastatic cascade and particularly impacts the interactions between tumor cells and host stroma at both primary and secondary sites. Here we review current evidence for a hypoxia-induced tumor secretome and its impact on metastatic progression. These studies have identified potential biomarkers and therapeutic targets that could be integrated into strategies for preventing and treating metastatic disease.
Collapse
Affiliation(s)
- Ying Liu
- The Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Gabrielle E Ciotti
- The Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - T S Karin Eisinger-Mathason
- The Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
32
|
Imakawa K, Bai R, Kusama K. Integration of molecules to construct the processes of conceptus implantation to the maternal endometrium. J Anim Sci 2018; 96:3009-3021. [PMID: 29554266 DOI: 10.1093/jas/sky103] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 03/15/2018] [Indexed: 12/22/2022] Open
Abstract
During the peri-implantation period, ruminant conceptuses go through rapid elongation, followed by their attachment to the uterine endometrial epithelial cells, during which interferon-tau (IFNT), a trophectodermal cytokine required for the process of maternal recognition of pregnancy, is expressed in a temporal and spatial manner. On day 22 (day 0 = day of estrus), 2 to 3 d after the initiation of bovine conceptus attachment to the uterine epithelium, when IFNT production begins to subside, the expression of molecules related to epithelial-mesenchymal transition, zinc finger E-box binding homeobox 1, snail family transcriptional repressor 2, N-cadherin, and vimentin was found in the trophectoderm. Through the use of in vitro coculture system with bovine trophoblast CT-1 and endometrial epithelial cells, a series of experiments have been conducted to elucidate mechanisms associated with the regulation of IFNT gene transcription and conceptus implantation, including epithelial-mesenchymal transition processes. Expression of IFNT, both up- and downregulation, during the peri-implantation period is tightly controlled. Cytokines and cell adhesion molecules such as epidermal growth factor, basic fibroblast growth factor, transforming growth factor beta, activin A, L-selectin-podocalyxin, and vascular cell adhesion molecule 1-integrin α4 expressed in utero all contribute to the initiation of epithelial-mesenchymal transition in the trophectoderm. These results indicate that conceptus implantation to the uterine endometrium proceeds while elongated conceptuses and endometria express cell adhesion molecules and their receptors, and the trophectoderm experiences epithelial-mesenchymal transition. Data accumulated suggest that while the conceptus and the endometrial epithelium adhere, trophectodermal cells must gain more flexibility for binucleate and possibly trinucleate cell formation during the peri-implantation period, and that understanding and constructing the conditions throughout implantation processes is key to improving ruminants' fertility.
Collapse
Affiliation(s)
- K Imakawa
- Animal Resource Science Center, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Ago, Kasama, Ibaraki, Japan
| | - R Bai
- Animal Resource Science Center, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Ago, Kasama, Ibaraki, Japan
| | - K Kusama
- Animal Resource Science Center, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Ago, Kasama, Ibaraki, Japan
| |
Collapse
|
33
|
Meurette O, Mehlen P. Notch Signaling in the Tumor Microenvironment. Cancer Cell 2018; 34:536-548. [PMID: 30146333 DOI: 10.1016/j.ccell.2018.07.009] [Citation(s) in RCA: 456] [Impact Index Per Article: 65.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/30/2018] [Accepted: 07/24/2018] [Indexed: 12/18/2022]
Abstract
The Notch signaling pathway regulates many aspects of cancer biology. Most attention has been given to its role in the transformed cell. However, it is now clear that cancer progression and metastasis depend on the bidirectional interactions between cancer cells and their environment, forming the tumor microenvironment (TME). These interactions are mediated and constantly evolve through paracrine and juxtacrine signaling. In this review, we discuss how Notch signaling takes an important part in regulating the crosstalk between the different compartments of the TME. We also address the consequences of the Notch-TME involvement from a therapeutic perspective.
Collapse
Affiliation(s)
- Olivier Meurette
- Apoptosis, Cancer and Development Laboratory- Equipe Labellisée 'La Ligue', LabEx DEVweCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, 69008 Lyon, France.
| | - Patrick Mehlen
- Apoptosis, Cancer and Development Laboratory- Equipe Labellisée 'La Ligue', LabEx DEVweCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, 69008 Lyon, France
| |
Collapse
|
34
|
Hamidi S, Sheng G. Epithelial-mesenchymal transition in haematopoietic stem cell development and homeostasis. J Biochem 2018; 164:265-275. [PMID: 30020470 DOI: 10.1093/jb/mvy063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/14/2018] [Indexed: 01/03/2025] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a morphogenetic process of cells that adopt an epithelial organization in their developmental ontogeny or homeostatic maintenance. Abnormalities in EMT regulation result in many malignant tumours in the human body. Tumours associated with the haematopoietic system, however, are traditionally not considered to involve EMT and haematopoietic stem cells (HSCs) are generally not associated with epithelial characteristics. In this review, we discuss the ontogeny and homeostasis of adult HSCs in the context of EMT intermediate states. We provide evidence that cell polarity regulation is critical for both HSC formation from embryonic dorsal aorta and HSC self-renewal and differentiation in adult bone marrow. HSC polarity is controlled by the same set of surface and transcriptional regulators as those described in canonical EMT processes. With an emphasis on partial EMT, we propose that the concept of EMT can be similarly applied in the study of HSC generation, maintenance and pathogenesis.
Collapse
Affiliation(s)
- Sofiane Hamidi
- Laboratory of Developmental Morphogenesis, International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Guojun Sheng
- Laboratory of Developmental Morphogenesis, International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
35
|
Kelaini S, Vilà-González M, Caines R, Campbell D, Eleftheriadou M, Tsifaki M, Magee C, Cochrane A, O'neill K, Yang C, Stitt AW, Zeng L, Grieve DJ, Margariti A. Follistatin-Like 3 Enhances the Function of Endothelial Cells Derived from Pluripotent Stem Cells by Facilitating β-Catenin Nuclear Translocation Through Inhibition of Glycogen Synthase Kinase-3β Activity. Stem Cells 2018; 36:1033-1044. [PMID: 29569797 PMCID: PMC6099345 DOI: 10.1002/stem.2820] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 02/10/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Abstract
The fight against vascular disease requires functional endothelial cells (ECs) which could be provided by differentiation of induced Pluripotent Stem Cells (iPS Cells) in great numbers for use in the clinic. However, the great promise of the generated ECs (iPS-ECs) in therapy is often restricted due to the challenge in iPS-ECs preserving their phenotype and function. We identified that Follistatin-Like 3 (FSTL3) is highly expressed in iPS-ECs, and, as such, we sought to clarify its possible role in retaining and improving iPS-ECs function and phenotype, which are crucial in increasing the cells' potential as a therapeutic tool. We overexpressed FSTL3 in iPS-ECs and found that FSTL3 could induce and enhance endothelial features by facilitating β-catenin nuclear translocation through inhibition of glycogen synthase kinase-3β activity and induction of Endothelin-1. The angiogenic potential of FSTL3 was also confirmed both in vitro and in vivo. When iPS-ECs overexpressing FSTL3 were subcutaneously injected in in vivo angiogenic model or intramuscularly injected in a hind limb ischemia NOD.CB17-Prkdcscid/NcrCrl SCID mice model, FSTL3 significantly induced angiogenesis and blood flow recovery, respectively. This study, for the first time, demonstrates that FSTL3 can greatly enhance the function and maturity of iPS-ECs. It advances our understanding of iPS-ECs and identifies a novel pathway that can be applied in cell therapy. These findings could therefore help improve efficiency and generation of therapeutically relevant numbers of ECs for use in patient-specific cell-based therapies. In addition, it can be particularly useful toward the treatment of vascular diseases instigated by EC dysfunction. Stem Cells 2018;36:1033-1044.
Collapse
Affiliation(s)
- Sophia Kelaini
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Marta Vilà-González
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Rachel Caines
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - David Campbell
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | | | - Marianna Tsifaki
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Corey Magee
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Amy Cochrane
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Karla O'neill
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Chunbo Yang
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Alan W Stitt
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Lingfang Zeng
- Cardiovascular Division, King's College London, London, United Kingdom
| | - David J Grieve
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Andriana Margariti
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
36
|
Xu L, Zheng L, Wang Z, Li C, Li S, Xia X, Zhang P, Li L, Zhang L. TNF-α-Induced SOX5 Upregulation Is Involved in the Osteogenic Differentiation of Human Bone Marrow Mesenchymal Stem Cells Through KLF4 Signal Pathway. Mol Cells 2018; 41:575-581. [PMID: 29890823 PMCID: PMC6030245 DOI: 10.14348/molcells.2018.2359] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/11/2018] [Accepted: 03/21/2018] [Indexed: 02/07/2023] Open
Abstract
Postmenopausal osteoporosis (PMOP) is a common systemic skeletal disease characterized by reduced bone mass and microarchitecture deterioration. Although differentially expressed SOX5 has been found in bone marrow from ovariectomized mice, its role in osteogenic differentiation in human mesenchymal stem cells (hMSCs) from bone marrow in PMOP remains unknown. In this study, we investigated the biological function of SOX5 and explore its molecular mechanism in hMSCs from patients with PMOP. Our findings showed that the mRNA and protein expression levels of SOX5 were upregulated in hMSCs isolated from bone marrow samples of PMOP patients. We also found that SOX5 overexpression decreased the alkaline phosphatase (ALP) activity and the gene expression of osteoblast markers including Collagen I, Runx2 and Osterix, which were increased by SOX5 knockdown using RNA interference. Furthermore, TNF-α notably upregulated the SOX5 mRNA expression level, and SOX5 knockdown reversed the effect of TNF-α on osteogenic differentiation of hMSCs. In addition, SOX5 overexpression increased Kruppel-like factor 4 (KLF4) gene expression, which was decreased by SOX5 silencing. KLF4 knockdown abrogated the suppressive effect of SOX5 overexpression on osteogenic differentiation of hMSCs. Taken together, our results indicated that TNF-α-induced SOX5 upregulation inhibited osteogenic differentiation of hMSCs through KLF4 signal pathway, suggesting that SOX5 might be a novel therapeutic target for PMOP treatment.
Collapse
Affiliation(s)
- Lijun Xu
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052,
China
| | - Lili Zheng
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052,
China
| | - Zhifang Wang
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052,
China
| | - Chong Li
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052,
China
| | - Shan Li
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052,
China
| | - Xuedi Xia
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052,
China
| | - Pengyan Zhang
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052,
China
| | - Li Li
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052,
China
| | - Lixia Zhang
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052,
China
| |
Collapse
|
37
|
Increasing aggressiveness of patient-derived xenograft models of cervix carcinoma during serial transplantation. Oncotarget 2018; 9:21036-21051. [PMID: 29765518 PMCID: PMC5940365 DOI: 10.18632/oncotarget.24783] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 02/27/2018] [Indexed: 12/13/2022] Open
Abstract
Four patient-derived xenograft (PDX) models (BK-12, ED-15, HL-16, LA-19) of carcinoma of the uterine cervix have been developed in our laboratory, and their stability during serial transplantation in vivo was investigated in this study. Two frozen cell stocks were established, one from xenografted tumors in passage 2 (early generation) and the other from xenografted tumors transplanted serially in mice for approximately two years (late generation), and the biology of late generation tumors was compared with that of early generation tumors. Late generation tumors showed higher incidence of lymph node metastases than early generation tumors in three models (ED-15, HL-16, LA-19), and the increased metastatic propensity was associated with increased tumor growth rate, increased microvascular density, and increased expression of angiogenesis-related and cancer stem cell-related genes. Furthermore, late generation tumors showed decreased fraction of pimonidazole-positive tissue (i.e., decreased fraction of hypoxic tissue) in two models (HL-16, LA-19) and decreased fraction of collagen-I-positive tissue (i.e., less extensive extracellular matrix) in two models (ED-15, HL-16). This study showed that serially transplanted PDXs may not necessarily mirror the donor patients’ diseases, and consequently, proper use of serially transplanted PDX models in translational cancer research requires careful molecular monitoring of the models.
Collapse
|
38
|
Mammoto T, Muyleart M, Konduri GG, Mammoto A. Twist1 in Hypoxia-induced Pulmonary Hypertension through Transforming Growth Factor-β-Smad Signaling. Am J Respir Cell Mol Biol 2018; 58:194-207. [PMID: 28915063 DOI: 10.1165/rcmb.2016-0323oc] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Pulmonary hypertension (PH) is a devastating pulmonary vascular disease characterized by aberrant muscularization of the normally nonmuscularized distal pulmonary arterioles. The expression of the transcription factor, Twist1, increases in the lungs of patients with pulmonary arterial hypertension. However, the mechanisms by which Twist1 controls the pathogenesis of PH remain unclear. It is becoming clear that endothelial-to-mesenchymal transition (EndMT) contributes to various vascular pathologies, including PH; Twist1 is known to mediate EndMT. In this report, we demonstrate that Twist1 overexpression increases transforming growth factor (TGF) β receptor2 (TGF-βR2) expression and Smad2 phosphorylation, and induces EndMT in cultured human pulmonary arterial endothelial (HPAE) cells, whereas a mutant construct of Twist1 at the serine 42 residue (Twist1S42A) fails to induce EndMT. We also implanted fibrin gel supplemented with HPAE cells on the mouse lung, and found that these HPAE cells form vascular structures and that Twist1-overexpressing HPAE cells undergo EndMT in the gel, whereas Twist1S42A-overexpressing cells do not. Furthermore, hypoxia-induced EndMT is inhibited in endothelial cells overexpressing Twist1S42A mutant construct in vitro. Hypoxia-induced accumulation of α-smooth muscle actin-positive cells in the pulmonary arterioles is attenuated in Tie2-specific Twist1 conditional knockout mice in vivo. These findings suggest that Twist1 serine 42 phosphorylation plays a key role in EndMT through TGF-β signaling and that modulation of Twist1 phosphorylation could be an effective strategy for managing PH.
Collapse
Affiliation(s)
- Tadanori Mammoto
- 1 Vascular Biology Program, Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts; and.,2 Department of Radiology and
| | - Megan Muyleart
- 2 Department of Radiology and.,3 Department of Pediatrics Medical College of Wisconsin, Milwaukee, Wisconsin
| | - G Ganesh Konduri
- 3 Department of Pediatrics Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Akiko Mammoto
- 1 Vascular Biology Program, Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts; and.,3 Department of Pediatrics Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
39
|
Sánchez-Duffhues G, García de Vinuesa A, Ten Dijke P. Endothelial-to-mesenchymal transition in cardiovascular diseases: Developmental signaling pathways gone awry. Dev Dyn 2017; 247:492-508. [PMID: 28891150 DOI: 10.1002/dvdy.24589] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/30/2017] [Accepted: 08/30/2017] [Indexed: 01/05/2023] Open
Abstract
The process named endothelial-to-mesenchymal transition (EndMT) was observed for the first time during the development of the chicken embryo several decades ago. Of interest, accumulating evidence suggests that EndMT plays a critical role in the onset and progression of multiple postnatal cardiovascular diseases. EndMT is controlled by a set of developmental signaling pathways, very similar to the process of epithelial-to-mesenchymal transition, which determine the activity of several EndMT transcriptional effectors. Once activated, these EndMT effectors regulate the expression of endothelial- and mesenchymal-specific genes, in part by interacting with specific motifs in promoter regions, eventually leading to the down-regulation of endothelial-specific features and acquisition of a fibroblast-like phenotype. Important technical advances in lineage tracing methods combined with experimental mouse models demonstrated the pathophysiological importance of EndMT for human diseases. In this review, we discuss the major signal transduction pathways involved in the activation and regulation of the EndMT program. Furthermore, we will review the latest discoveries on EndMT, focusing on cardiovascular diseases, and in particular on its role in vascular calcification, pulmonary arterial hypertension, and organ fibrosis. Developmental Dynamics 247:492-508, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Gonzalo Sánchez-Duffhues
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, The Netherlands
| | - Amaya García de Vinuesa
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, The Netherlands
| | - Peter Ten Dijke
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, The Netherlands
| |
Collapse
|
40
|
Zhao Z, Rahman MA, Chen ZG, Shin DM. Multiple biological functions of Twist1 in various cancers. Oncotarget 2017; 8:20380-20393. [PMID: 28099910 PMCID: PMC5386770 DOI: 10.18632/oncotarget.14608] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 01/01/2017] [Indexed: 01/05/2023] Open
Abstract
Twist1 is a well-known regulator of transcription during embryonic organogenesis in many species. In humans, Twist1 malfunction was first linked to Saethre-Chotzen syndrome and later identified to play an essential role in tumor initiation, stemness, angiogenesis, invasion, metastasis, and chemo-resistance in a variety of carcinomas, sarcomas, and hematological malignances. In this review, we will first focus on systematically elaborating the diverse pathological functions of Twist1 in various cancers, then delineating the intricate underlying network of molecular mechanisms, based on which we will summarize current therapeutic strategies in cancer treatment that target and modulate Twist1-involved signaling pathways. Most importantly, we will put special emphasis on revealing the independence and interdependency of these multiple biological functions of Twist1, piecing together the whole delicate picture of Twist1's diversified pathological roles in different cancers and providing new perspectives to guide future research.
Collapse
Affiliation(s)
- Zhixiang Zhao
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, United States of America.,Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mohammad Aminur Rahman
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, United States of America
| | - Zhuo G Chen
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, United States of America
| | - Dong M Shin
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, United States of America
| |
Collapse
|
41
|
Liu Z, Qi L, Li Y, Zhao X, Sun B. VEGFR2 regulates endothelial differentiation of colon cancer cells. BMC Cancer 2017; 17:593. [PMID: 28854900 PMCID: PMC5577671 DOI: 10.1186/s12885-017-3578-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 08/22/2017] [Indexed: 12/13/2022] Open
Abstract
Background Recent studies suggested that cancer stem-like cells contribute to tumor vasculogenesis by differentiating into endothelial cells. However, such process is governed by still undefined mechanism. Methods At varying differentiation levels, three representative colon cancer cells were cultured in endothelial-inducing conditioned medium: human colon cancer cells HCT116 (HCT116) (poorly differentiated), SW480 (moderately differentiated), and HT29 (well differentiated). We tested for expression of endothelial markers (cluster of differentiation (CD) 31, CD34, and vascular endothelial (VE)-cadherin and their ability to form tube-like structures in 3D culture. We also observed VEGF secretion and expressions of endothelial markers and VEGFRs in HCT116 cells under hypoxia to simulate physiological conditions. In in vitro and in xenotransplantation experiments, VE growth factor receptor 2 (VEGFR2) antagonist SKLB1002 was used to test effect of VEGFR2 in endothelial differentiation of HCT116 cells. Expression levels of VEGFR2 and VE-cadherin were assessed by immunohistochemistry of human colon cancer tissues to evaluate clinicopathological significance of VEGFR2. Results After culturing in endothelial-inducing conditioned medium, poorly differentiated HCT116 cells expressed endothelial markers and formed tube-like structure in vitro. HCT116 cells secreted more endogenous VEGF and expressed higher VEGFR2 under hypoxia. SKLB1002 impaired endothelial differentiation in vitro and xenotransplantation experiments, suggesting a VEGFR2-dependent mechanism. Increased expression of VEGFR2 correlated with differentiation, metastasis/recurrence, and poor prognosis in 203 human colon cancer samples. Positive correlation was observed between VEGFR2 and VE-cadherin expression. Conclusions VEGFR2 regulates endothelial differentiation of colon cancer cell and may be potential platform for anti-angiogenesis cancer therapy.
Collapse
Affiliation(s)
- Zhiyong Liu
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.,The Key Laboratory of Tianjin Cancer Prevention and Treatment, Tianjin, 300060, China.,National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Lisha Qi
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.,The Key Laboratory of Tianjin Cancer Prevention and Treatment, Tianjin, 300060, China.,National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yixian Li
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Xiulan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Baocun Sun
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China. .,Department of Pathology, Tianjin Medical University, Tianjin, 300070, China. .,The Key Laboratory of Tianjin Cancer Prevention and Treatment, Tianjin, 300060, China. .,National Clinical Research Center for Cancer, Tianjin, 300060, China.
| |
Collapse
|
42
|
Chen Z, Xu D, Zhang T. Inhibition of proliferation and invasion of hepatocellular carcinoma cells by lncRNA-ASLNC02525 silencing and the mechanism. Int J Oncol 2017; 51:851-858. [PMID: 28713968 DOI: 10.3892/ijo.2017.4069] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 06/26/2017] [Indexed: 11/06/2022] Open
Abstract
One of the most differentially expressed long non-coding RNAs (lncRNAs) that we identified by high throughput screening from liver cancer and para-carcinoma tissues, ASLNC02525, was highly expressed in the tissues and cell lines of liver cancer but not in adjacent tissues or normal hepatic cells. Knockdown of ASLNC02525 in hepatocellular carcinoma cells inhibited the proliferation and invasion. In the process, expression level of transcription factor twist1 (twist‑related protein 1) was reduced, but no change at transcription level was observed. According to bioinformatics analysis, ASLNC02525 may play a crucial role in inactivation of regulation of twist1 by hsa-miRNA-489-3p. The mechanism study revealed that ASLNC02525, as an RNA sponge, broke the negative regulation of twist1 by hsa-miRNA-489-3p, and once ASLNC02525 was silenced, the highly expressed hsa-miRNA‑489-3p regained its regulation on twist1 and inhibited the proliferation and invasion. The importance of this study lies in shedding light on the potential for lncRNAs to become targets for gene therapy, by demonstrating that lncRNAs can suppress tumor inhibiting activity of miRNAs via breaking regulation of some miRNA target genes.
Collapse
Affiliation(s)
- Zi Chen
- Department of Hematology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Dongwen Xu
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Tao Zhang
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
43
|
Imakawa K, Bai R, Fujiwara H, Ideta A, Aoyagi Y, Kusama K. Continuous model of conceptus implantation to the maternal endometrium. J Endocrinol 2017; 233:R53-R65. [PMID: 28213399 DOI: 10.1530/joe-16-0490] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 02/17/2017] [Indexed: 12/17/2022]
Abstract
As placental morphology as well as trophoblast characteristics exhibit wide diversity across mammalian species, underling molecules were also thought to vary greatly. In the majority of cases, however, regardless of the mode of implantation, physiological and biochemical processes in conceptus implantation to the maternal endometrium including the kinds of gene expression and their products are now considered to share many similarities. In fact, recent progress has identified that in addition to the hormones, cytokines, proteases and cell adhesion molecules classically characterized, molecules related to lymphocyte homing and epithelial-mesenchymal transition (EMT) are all required for the progression of conceptus implantation to placentation. In this review, therefore, the newest findings are all incorporated into the molecular and cellular events related to conceptus implantation to the maternal endometrium; primarily from non-invasive bovine placentation and also from invasive human implantation.
Collapse
Affiliation(s)
- Kazuhiko Imakawa
- Animal Resource Science CenterGraduate School of Agricultural and Life Sciences, The University of Tokyo, Kasama, Ibaraki, Japan
| | - Rulan Bai
- Animal Resource Science CenterGraduate School of Agricultural and Life Sciences, The University of Tokyo, Kasama, Ibaraki, Japan
| | - Hiroshi Fujiwara
- Faculty of MedicineInstitute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa, Japan
| | - Atsushi Ideta
- Zennoh Embryo Transfer CenterKamishihoro, Hokkaido, Japan
| | - Yoshito Aoyagi
- Zennoh Embryo Transfer CenterKamishihoro, Hokkaido, Japan
| | - Kazuya Kusama
- Animal Resource Science CenterGraduate School of Agricultural and Life Sciences, The University of Tokyo, Kasama, Ibaraki, Japan
| |
Collapse
|
44
|
Liu S, Yang H, Chen Y, He B, Chen Q. Krüppel-Like Factor 4 Enhances Sensitivity of Cisplatin to Lung Cancer Cells and Inhibits Regulating Epithelial-to-Mesenchymal Transition. Oncol Res 2017; 24:81-7. [PMID: 27296948 PMCID: PMC7838665 DOI: 10.3727/096504016x14597766487717] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In order to improve therapeutic efficacy, it is a current emergency to better know the mechanisms underlying cisplatin resistance in lung cancer cells. In this study, we aim to investigate the role of Krüppel-like factor 4 (KLF4) in cisplatin-resistant lung cancer cells. We developed cisplatin-resistant lung cancer cell line A549/DDP, and then a battery of experiments was used to analyze the effects of KLF4 in cisplatin resistance of lung cancer. We found that KLF4 was significantly downregulated in cisplatin-resistant A549 cells and forced KLF4 expression inhibited cell growth and induced apoptosis. Further, we found that overexpression of KLF4 was able to inhibit cell migration and invasion, to inhibit the expression of Slug, Twist, and vimentin, and to increase the expression of E-cadherin and subsequent inhibition of the EMT process. Thus, overexpression of KLF4 may be a potential strategy for lung cancer treatment, especially for cisplatin-resistant cases.
Collapse
Affiliation(s)
- Shenggang Liu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | | | | | | | | |
Collapse
|
45
|
Xu Y, Qin L, Sun T, Wu H, He T, Yang Z, Mo Q, Liao L, Xu J. Twist1 promotes breast cancer invasion and metastasis by silencing Foxa1 expression. Oncogene 2017; 36:1157-1166. [PMID: 27524420 PMCID: PMC5311074 DOI: 10.1038/onc.2016.286] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 06/12/2016] [Accepted: 07/05/2016] [Indexed: 12/31/2022]
Abstract
The heterogeneous breast cancers can be classified into different subtypes according to their histopathological characteristics and molecular signatures. Foxa1 expression is linked with luminal breast cancer (LBC) with good prognosis, whereas Twist1 expression is associated with basal-like breast cancer (BLBC) with poor prognosis owing to its role in promoting epithelial-to-mesenchymal transition (EMT), invasiveness and metastasis. However, the regulatory and functional relationships between Twist1 and Foxa1 in breast cancer progression are unknown. In this study, we demonstrate that in the estrogen receptor (ERα)-positive LBC cells Twist1 silences Foxa1 expression, which has an essential role in relieving Foxa1-arrested migration, invasion and metastasis of breast cancer cells. Mechanistically, Twist1 binds to Foxa1 proximal promoter and recruits the NuRD transcriptional repressor complex to de-acetylate H3K9 and repress RNA polymerase II recruitment. Twist1 also silences Foxa1 promoter by inhibiting AP-1 recruitment. Twist1 expression in MCF7 cells silenced Foxa1 expression, which was concurrent with the induction of EMT, migration, invasion and metastasis of these cells. Importantly, restored Foxa1 expression in these cells largely inhibited Twist1-promoted migration, invasion and metastasis. Restored Foxa1 expression did not change the Twist1-induced mesenchymal cellular morphology and the expression of Twist1-regulated E-cadherin, β-catenin, vimentin and Slug, but it partially rescued Twist1-silenced ERα and cytokeratin 8 expression and reduced Twist1-induced integrin α5, integrin β1 and MMP9 expression. In a xenografted mouse model, restored Foxa1 also increased Twist1-repressed LBC markers and decreased Twist1-induced BLBC markers. Furthermore, Twist1 expression is negatively correlated with Foxa1 in the human breast tumors. The tumors with high Twist1 and low Foxa1 expressions are associated with poor distant metastasis-free survival. These results demonstrate that Twist1's silencing effect on Foxa1 expression is largely responsible for Twist1-induced migration, invasion and metastasis, but less responsible for Twist1-induced mesenchymal morphogenesis and expression of certain EMT markers.
Collapse
Affiliation(s)
- Yixiang Xu
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas
| | - Li Qin
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Tong Sun
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Hongmei Wu
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Tao He
- Institution for Cancer Medicine and College of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Zhihui Yang
- Institution for Cancer Medicine and College of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Qianxing Mo
- Dan L. Duncan Cancer Center and Department of Medicine-Hematology/Oncology, Baylor College of Medicine, Houston, Texas
| | - Lan Liao
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Jianming Xu
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
- Institution for Cancer Medicine and College of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
46
|
Pei H, Li Y, Liu M, Chen Y. Targeting Twist expression with small molecules. MEDCHEMCOMM 2016; 8:268-275. [PMID: 30108743 DOI: 10.1039/c6md00561f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/01/2016] [Indexed: 12/14/2022]
Abstract
Twist, as one of the important embryonic transcription factors, regulates epithelial-mesenchymal transition (EMT) and migration in embryo formation and cancer development. Both Twist-1 and Twist-2 are rarely detectable in healthy adult tissues, but are frequently overexpressed in multiple kinds of human cancer tissues, such as breast, prostate, uterus, liver, melanoma, etc. Twist is considered as a crucial EMT inductor and correlated with carcinoma aggression, invasion and metastasis. In the past decades, in-depth investigation has been reported in terms of the role of Twist in cancers; in addition, several kinds of small molecules have played important roles in studying the effect of Twist on cancer development, suggesting that Twist can be regarded as one of the important potential targets for cancer treatment. Hence we provide a brief overview of Twist and several small molecules targeting its expression, highlighting the biological features that make it a charming target for cancer therapy.
Collapse
Affiliation(s)
- Haixiang Pei
- Shanghai Key Laboratory of Regulatory Biology , The Institute of Biomedical Sciences and School of Life Sciences , East China Normal University , Shanghai , 200241 , China . ; ; Tel: +86 21 2420 6647
| | - Yunqi Li
- Shanghai Key Laboratory of Regulatory Biology , The Institute of Biomedical Sciences and School of Life Sciences , East China Normal University , Shanghai , 200241 , China . ; ; Tel: +86 21 2420 6647
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology , The Institute of Biomedical Sciences and School of Life Sciences , East China Normal University , Shanghai , 200241 , China . ; ; Tel: +86 21 2420 6647
| | - Yihua Chen
- Shanghai Key Laboratory of Regulatory Biology , The Institute of Biomedical Sciences and School of Life Sciences , East China Normal University , Shanghai , 200241 , China . ; ; Tel: +86 21 2420 6647
| |
Collapse
|
47
|
Lee JY, Kong G. Roles and epigenetic regulation of epithelial-mesenchymal transition and its transcription factors in cancer initiation and progression. Cell Mol Life Sci 2016; 73:4643-4660. [PMID: 27460000 PMCID: PMC11108467 DOI: 10.1007/s00018-016-2313-z] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/15/2016] [Accepted: 07/19/2016] [Indexed: 02/07/2023]
Abstract
The epithelial-mesenchymal transition (EMT) is a crucial developmental process by which epithelial cells undergo a mesenchymal phenotypic change. During EMT, epigenetic mechanisms including DNA methylation and histone modifications are involved in the regulation of EMT-related genes. The epigenetic gene silencing of the epithelial marker E-cadherin has been well characterized. In particular, three major transcriptional repressors of E-cadherin, Snail, ZEB, and Twist families, also known as EMT-inducing transcription factors (EMT-TFs), play a crucial role in this process by cooperating with multiple epigenetic modifiers. Furthermore, recent studies have identified the novel epigenetic modifiers that control the expression of EMT-TFs, and these modifiers have emerged as critical regulators of cancer development and as novel therapeutic targets for human cancer. In this review, the diverse functions of EMT-TFs in cancer progression, the cooperative mechanisms of EMT-TFs with epigenetic modifiers, and epigenetic regulatory roles for the expression of EMT-TFs will be discussed.
Collapse
Affiliation(s)
- Jeong-Yeon Lee
- Institute for Bioengineering and Biopharmaceutical Research (IBBR), Hanyang University, Seoul, Republic of Korea
| | - Gu Kong
- Department of Pathology, College of Medicine, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea.
| |
Collapse
|
48
|
Kusama K, Bai R, Ideta A, Aoyagi Y, Okuda K, Imakawa K. Regulation of epithelial to mesenchymal transition in bovine conceptuses through the interaction between follistatin and activin A. Mol Cell Endocrinol 2016; 434:81-92. [PMID: 27321969 DOI: 10.1016/j.mce.2016.06.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/13/2016] [Accepted: 06/14/2016] [Indexed: 11/25/2022]
Abstract
Dynamic changes in bovine conceptus and endometrium occur during early gestation, in which the conceptus undergoes epithelial to mesenchymal transition (EMT) after the conceptus attachment to endometrium. To characterize EMT inducing factors, we initially undertook iTRAQ analysis with bovine uterine flushing (UF) obtained from pregnant animals on days 17 (P17: pre-attachment) and 20 (P20: post-attachment). The iTRAQ analysis demonstrated that follistatin (FST), an inhibitor of activin A, increased in P20 UF. We then found that FST decreased in P22 conceptuses, whereas elevated activin A found in P20 UF and endometria was further increased on P22. In addition, phosphorylated SMAD2 increased in P22 conceptuses. In bovine trophoblast cells, the treatment with P22 UF or activin An up-regulated EMT marker expressions, which were inhibited by FST. These results suggest that the initiation of bovine conceptus EMT could be regulated through the spatiotemporal expression of FST or activin A during the peri-attachment period.
Collapse
Affiliation(s)
- Kazuya Kusama
- Laboratory of Theriogenology and Animal Breeding, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan
| | - Rulan Bai
- Laboratory of Theriogenology and Animal Breeding, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan
| | - Atsushi Ideta
- Zennoh Embryo Transfer Center, Hokkaido, 080-1407, Japan
| | - Yoshito Aoyagi
- Zennoh Embryo Transfer Center, Hokkaido, 080-1407, Japan
| | - Kiyoshi Okuda
- Laboratory of Reproductive Endocrinology, Graduate School of Natural Science and Technology, Okayama University, Okayama, 700-8530, Japan
| | - Kazuhiko Imakawa
- Laboratory of Theriogenology and Animal Breeding, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan; Animal Resource Science Center, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Ibaraki, 319-0206, Japan.
| |
Collapse
|
49
|
Ping YF, Zhang X, Bian XW. Cancer stem cells and their vascular niche: Do they benefit from each other? Cancer Lett 2016; 380:561-567. [DOI: 10.1016/j.canlet.2015.05.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 04/07/2015] [Accepted: 05/08/2015] [Indexed: 12/22/2022]
|
50
|
Vascular Transdifferentiation in the CNS: A Focus on Neural and Glioblastoma Stem-Like Cells. Stem Cells Int 2016; 2016:2759403. [PMID: 27738435 PMCID: PMC5055959 DOI: 10.1155/2016/2759403] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 09/05/2016] [Indexed: 01/12/2023] Open
Abstract
Glioblastomas are devastating and extensively vascularized brain tumors from which glioblastoma stem-like cells (GSCs) have been isolated by many groups. These cells have a high tumorigenic potential and the capacity to generate heterogeneous phenotypes. There is growing evidence to support the possibility that these cells are derived from the accumulation of mutations in adult neural stem cells (NSCs) as well as in oligodendrocyte progenitors. It was recently reported that GSCs could transdifferentiate into endothelial-like and pericyte-like cells both in vitro and in vivo, notably under the influence of Notch and TGFβ signaling pathways. Vascular cells derived from GBM cells were also observed directly in patient samples. These results could lead to new directions for designing original therapeutic approaches against GBM neovascularization but this specific reprogramming requires further molecular investigations. Transdifferentiation of nontumoral neural stem cells into vascular cells has also been described and conversely vascular cells may generate neural stem cells. In this review, we present and discuss these recent data. As some of them appear controversial, further validation will be needed using new technical approaches such as high throughput profiling and functional analyses to avoid experimental pitfalls and misinterpretations.
Collapse
|