106 results on '"Tumor initiating cell"'
Search Results
2. Re-Recognizing the Cellular Origin of the Primary Epithelial Tumors of the Liver
- Author
-
Feng J, Zhu R, Yin Y, Wang S, Zhou L, Lv F, and Zhao D
- Subjects
hcc ,tumor initiating cell ,hepatic progenitor ,neuroendocrine tumors ,hepatoblastoma ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Jiliang Feng,1 Ruidong Zhu,2 Yu Yin,3 Shanshan Wang,1 Lei Zhou,4 Fudong Lv,1 Dawei Zhao5 1Clinical-Pathology Center, Beijing You-An Hospital, Capital Medical University, Beijing, 100069, People’s Republic of China; 2General Surgical Center, Beijing You-An Hospital, Capital Medical University, Beijing, 100069, People’s Republic of China; 3Department of Pathology, Anhui Medical University, Hefei, 230032, People’s Republic of China; 4Department of Pathology, First Affiliated Hospital of Bengbu Medical College/Bengbu Medical College, Bengbu, 233004, People’s Republic of China; 5Department of Medical Imaging, Capital Medical University, Beijing, 100069, People’s Republic of ChinaCorrespondence: Jiliang FengClinical-Pathology Center, Beijing You-An Hospital, Capital Medical University, No. 8, Xitoutiao, Youanmenwai Street, FengTai District, Beijing, 100069, People’s Republic of ChinaTel +86-10-83997342Fax +86-10-83997343Email jiliangfeng@ccmu.edu.cnAbstract: The primary epithelial tumors of the liver (PETL) are composed of a series of heterogeneous tumors. Although the classification of PETLs has been updated several times by the World Health Organization, the cellular origins of some tumors in this family remain to be precisely depicted. In addition, certain tumors in different categories have similar histology, molecular phenotypes and biological characteristics, suggesting that they may have the same cellular origin. In this work, a narrative review method was adopted to review the relevant papers. By comparing the expression profiles of biomarkers of liver epithelium at different lineages and stages of differentiation, the cells-of-origin of some major members of the PETL family were reassessed. We propose that 1) hepatic adenomas, hepatocellular carcinomas (HCCs) and pure fetal hepatoblastomas (HBs) share the same spectrum in their cellular origin including the hepatocytic-committed progenitors (HCP) and their differentiated descendants. 2) Bile duct adenomas, peribiliary cysts and intrahepatic cholangiocellular carcinomas (ICCs) can share the same spectrum in their cellular origin including the cholangiocytic-committed progenitors (CCP) and their differentiated descendants. 3) The cells-of-origin of embryonal HBs include liver stem cells (LSCs), hepatoblasts, and transitional cells between them. Embryonal HB with small cell element, small cell undifferentiated HB and small cell neuroendocrine carcinoma of the liver can have the same or similar cells-of-origin from LSC. Embryonal HB lacking the small cell component of the LSC phenotype and presenting both hepatocytic and bile duct/ductule components may originate from actual hepatoblasts/hepatic progenitor cells (HPCs) as the combined HCC-ICC does. 4) Teratoid hepatoblastoma and mixed epithelial/mesenchymal HBs can be derived from the LSCs or even less committed extrahepatic pluripotent stem cell. 5) Many members of the PETLs family, including those derived from LSCs, hepatoblasts/HPCs, early HCPs and CCPs, have neuroendocrine potentiality. Except for those primary hepatic neuroendocrine tumor (PHNET) exhibit hepatocytic and/or cholangiocytic phenotypes, other PHNETs subtype may be derived from the descendants of LSC that differentiate towards the upper digestive tract, pancreas or other lineages.Keywords: HCC, tumor-initiating cell, hepatic progenitor, neuroendocrine tumors, hepatoblastoma
- Published
- 2021
3. HDAC Class I Inhibitor Domatinostat Preferentially Targets Glioma Stem Cells over Their Differentiated Progeny.
- Author
-
Nakagawa-Saito, Yurika, Saitoh, Shinichi, Mitobe, Yuta, Sugai, Asuka, Togashi, Keita, Suzuki, Shuhei, Kitanaka, Chifumi, and Okada, Masashi
- Subjects
- *
STEM cells , *CANCER stem cells , *GLIOMAS , *BRAIN tumors , *CELL death , *CANCER cells - Abstract
Cancer stem cells (CSCs) are in general characterized by higher resistance to cell death and cancer therapies than non-stem differentiated cancer cells. However, we and others have recently revealed using glioma stem cells (GSCs) as a model that, unexpectedly, CSCs have specific vulnerabilities that make them more sensitive to certain drugs compared with their differentiated counterparts. We aimed in this study to discover novel drugs targeting such Achilles' heels of GSCs as anti-GSC drug candidates to be used for the treatment of glioblastoma, the most therapy-resistant form of brain tumors. Here we report that domatinostat (4SC-202), a class I HDAC inhibitor, is one such candidate. At concentrations where it showed no or minimal growth inhibitory effect on differentiated GSCs and normal cells, domatinostat effectively inhibited the growth of GSCs mainly by inducing apoptosis. Furthermore, GSCs that survived domatinostat treatment lost their self-renewal capacity. These results suggested that domatinostat is a unique drug that selectively eliminates GSCs not only physically by inducing cell death but also functionally by inhibiting their self-renewal. Our findings also imply that class I HDACs and/or LSD1, another target of domatinostat, may possibly have a specific role in the maintenance of GSCs and therefore could be an attractive target in the development of anti-GSC therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
4. CBP-mediated Wnt3a/β-catenin signaling promotes cervical oncogenesis initiated by Piwil2
- Author
-
Dingqing Feng, Keqin Yan, Haiyan Liang, Jing Liang, Wenhui Wang, Huan Yu, Ying Zhou, Weidong Zhao, Zhongjun Dong, and Bin Ling
- Subjects
Cervical cancer ,Piwil2 ,Wnt signaling ,Tumor initiating cell ,CBP ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Our previous work demonstrated that Piwil2 reactivated by the human papillomavirus oncoproteins E6 and E7 may reprogram somatic cells into tumor-initiating cells (TICs), which contribute to cervical neoplasia lesions. Maintaining the stemness of TICs is critical for the progression of cervical lesions. Here, we determined that canonical Wnt signaling was aberrantly activated in HaCaT cells transfected with lentivirus expressing Piwil2 and in cervical lesion specimens of low-grade squamous intraepithelial lesion, high-grade squamous intraepithelial lesion, and invasive carcinoma. Blocking the β-catenin and CREB binding protein interaction with ICG-001 significantly downregulated the reprogramming factors c-Myc, Nanog, Oct4, Sox2, and Klf4, thus leading to cell differentiation and preventing tumorigenicity in Piwil2-overexpressing HaCaT cells. Similarly, Piwil2 also critically regulated the canonical Wnt signaling pathway in cervical cancer. We further demonstrated that ICG-001 increased cisplatin sensitivity and significantly suppressed tumor growth of cervical cancer alone or in combination with cisplatin both in vitro and in vivo. The β-catenin/ CREB binding protein-mediated transcription activated by Piwil2 is essential for the maintenance of TICs, therefore contributing to the progression of cervical oncogenesis.
- Published
- 2021
- Full Text
- View/download PDF
5. Reactive oxygen species produced by altered tumor metabolism impacts cancer stem cell maintenance
- Author
-
Kaysaw Tuy, Lucas Rickenbacker, and Anita B. Hjelmeland
- Subjects
Cancer stem cell ,Tumor initiating cell ,Metabolism ,Reactive oxygen species ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Controlling reactive oxygen species (ROS) at sustainable levels can drive multiple facets of tumor biology, including within the cancer stem cell (CSC) population. Tight regulation of ROS is one key component in CSCs that drives disease recurrence, cell signaling, and therapeutic resistance. While ROS are well-appreciated to need oxygen and are a product of oxidative phosphorylation, there are also important roles for ROS under hypoxia. As hypoxia promotes and sustains major stemness pathways, further consideration of ROS impacts on CSCs in the tumor microenvironment is important. Furthermore, glycolytic shifts that occur in cancer and may be promoted by hypoxia are associated with multiple mechanisms to mitigate oxidative stress. This altered metabolism provides survival advantages that sustain malignant features, such as proliferation and self-renewal, while producing the necessary antioxidants that reduce damage from oxidative stress. Finally, disease recurrence is believed to be attributed to therapy resistant CSCs which can be quiescent and have changes in redox status. Effective DNA damage response pathways and/or a slow-cycling state can protect CSCs from the genomic catastrophe induced by irradiation and genotoxic agents. This review will explore the delicate, yet complex, relationship between ROS and its pleiotropic role in modulating the CSC.
- Published
- 2021
- Full Text
- View/download PDF
6. HDAC Class I Inhibitor Domatinostat Preferentially Targets Glioma Stem Cells over Their Differentiated Progeny
- Author
-
Yurika Nakagawa-Saito, Shinichi Saitoh, Yuta Mitobe, Asuka Sugai, Keita Togashi, Shuhei Suzuki, Chifumi Kitanaka, and Masashi Okada
- Subjects
glioma initiating cell ,tumor initiating cell ,epigenetic modulation ,stemness ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Cancer stem cells (CSCs) are in general characterized by higher resistance to cell death and cancer therapies than non-stem differentiated cancer cells. However, we and others have recently revealed using glioma stem cells (GSCs) as a model that, unexpectedly, CSCs have specific vulnerabilities that make them more sensitive to certain drugs compared with their differentiated counterparts. We aimed in this study to discover novel drugs targeting such Achilles’ heels of GSCs as anti-GSC drug candidates to be used for the treatment of glioblastoma, the most therapy-resistant form of brain tumors. Here we report that domatinostat (4SC-202), a class I HDAC inhibitor, is one such candidate. At concentrations where it showed no or minimal growth inhibitory effect on differentiated GSCs and normal cells, domatinostat effectively inhibited the growth of GSCs mainly by inducing apoptosis. Furthermore, GSCs that survived domatinostat treatment lost their self-renewal capacity. These results suggested that domatinostat is a unique drug that selectively eliminates GSCs not only physically by inducing cell death but also functionally by inhibiting their self-renewal. Our findings also imply that class I HDACs and/or LSD1, another target of domatinostat, may possibly have a specific role in the maintenance of GSCs and therefore could be an attractive target in the development of anti-GSC therapies.
- Published
- 2022
- Full Text
- View/download PDF
7. mTOR Inhibition Ablates Cisplatin-Resistant Salivary Gland Cancer Stem Cells.
- Author
-
Nakano, T., Warner, K.A., Oklejas, A.E., Zhang, Z., Rodriguez-Ramirez, C., Shuman, A.G., and Nör, J.E.
- Subjects
MTOR inhibitors ,CISPLATIN ,SALIVARY gland cancer ,CANCER stem cells ,DRUG resistance in cancer cells ,HEAD & neck cancer - Abstract
Patients with advanced salivary gland mucoepidermoid carcinoma (MEC) are treated with surgery and radiotherapy, as current systemic therapies are largely ineffective. As such, current treatment frequently leads to poor long-term survival due to locoregional recurrence or metastases. We have shown that salivary gland cancer stem cells (CSCs) are resistant to platinum-based chemotherapy and drive tumor progression. The purpose of this study was to investigate the effect of therapeutic inhibition of mTOR (mechanistic target of rapamycin) on resistance of CSCs to cisplatin, a prototypic platinum-based chemotherapeutic agent. Viability assays determined the effect of several inhibitors of PI3k/mTOR signaling (e.g., temsirolimus, BKM120, AZD8055, PF4708671) and/or cisplatin on survival of human MEC cells. The impact of mTOR inhibitors and/or cisplatin on MEC stemness was examined with salisphere assays, flow cytometry for ALDH/CD44 (CSC markers for MEC), and Western blots for Bmi-1 expression (marker of stem cell self-renewal). Salivary gland MEC patient-derived xenografts were used to examine the effect of cisplatin and/or temsirolimus on CSCs in vivo. We observed that cisplatin induced mTOR and S6K1 phosphorylation, increased the number and size of MEC salispheres, and induced Bmi-1 expression and the fraction of CSCs in MEC models in vitro. Cisplatin also increased the fraction of CSCs in vivo. In contrast, mTOR inhibition (e.g., temsirolimus) blocked cisplatin-induced Bmi-1 expression and salisphere formation in vitro. Remarkably, temsirolimus slowed down tumor growth and decreased the fraction of CSCs (P < 0.05) even in presence of cisplatin in a short-term in vivo experiment. Collectively, these results demonstrate that therapeutic inhibition of mTOR ablates cytotoxic-resistant CSCs, and they suggest that a combination of an mTOR inhibitor and platinum-based chemotherapy might be beneficial to patients with salivary gland mucoepidermoid carcinoma. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
8. CBP-mediated Wnt3a/β-catenin signaling promotes cervical oncogenesis initiated by Piwil2.
- Author
-
Feng, Dingqing, Yan, Keqin, Liang, Haiyan, Liang, Jing, Wang, Wenhui, Yu, Huan, Zhou, Ying, Zhao, Weidong, Dong, Zhongjun, and Ling, Bin
- Subjects
- *
CERVICAL intraepithelial neoplasia , *NEOPLASTIC cell transformation , *WNT signal transduction , *CREB protein , *SOMATIC cells , *CERVICAL cancer - Abstract
Our previous work demonstrated that Piwil2 reactivated by the human papillomavirus oncoproteins E6 and E7 may reprogram somatic cells into tumor-initiating cells (TICs), which contribute to cervical neoplasia lesions. Maintaining the stemness of TICs is critical for the progression of cervical lesions. Here, we determined that canonical Wnt signaling was aberrantly activated in HaCaT cells transfected with lentivirus expressing Piwil2 and in cervical lesion specimens of low-grade squamous intraepithelial lesion, high-grade squamous intraepithelial lesion, and invasive carcinoma. Blocking the β-catenin and CREB binding protein interaction with ICG-001 significantly downregulated the reprogramming factors c-Myc, Nanog, Oct4, Sox2 , and Klf4 , thus leading to cell differentiation and preventing tumorigenicity in Piwil2-overexpressing HaCaT cells. Similarly, Piwil2 also critically regulated the canonical Wnt signaling pathway in cervical cancer. We further demonstrated that ICG-001 increased cisplatin sensitivity and significantly suppressed tumor growth of cervical cancer alone or in combination with cisplatin both in vitro and in vivo. The β-catenin/ CREB binding protein-mediated transcription activated by Piwil2 is essential for the maintenance of TICs, therefore contributing to the progression of cervical oncogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
9. Kinomic profiling of glioblastoma cells reveals PLCG1 as a target in restricted glucose
- Author
-
Kiera Walker, Nathaniel H. Boyd, Joshua C. Anderson, Christopher D. Willey, and Anita B. Hjelmeland
- Subjects
Glioblastoma ,Tumor initiating cell ,Cancer stem cell ,PLCG1 ,Tumor microenvironment ,Restricted glucose ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Abstract Background For glioblastoma (GBM) treatments to be effective in vivo, understanding the effects of the tumor microenvironment is imperative. In traditional cell culture conditions, glucose concentrations do not model physiologic levels, nor the diminished concentrations found in tumor niches. We therefore sought to profile the differences in kinase activity in GBM cells cultured in restricted glucose to identify pathways that could be targeted with small molecule inhibitors. Methods Using the PamStation12 platform, we examined the ability of GBM lysates from cells cultured in standard or low glucose conditions to phosphorylate 144 tyrosine and 144 serine/threonine peptides that correspond to known protein phosphorylation sites. Potential kinase targets were identified and validated using small molecule kinase inhibitors in GBM spheroid cultures. Results Using results from two GBM patient-derived xenografts, we determined common changes to peptides derived from Phospholipase C, Gamma 1 (PLCG1) and Raf-1. Using PLC and Raf inhibitors, we found a significantly stronger growth inhibitory effect of the PLC inhibitor U73122 under restricted glucose conditions. In contrast, Raf inhibitors were significantly growth inhibitory regardless of the nutrient level tested. Conclusions Together, our data demonstrate that kinase activity is altered in low glucose conditions and that kinomic profiling can assist with the identification of effective strategies to target GBM growth. Our data further suggest the importance of accurately modeling the tumor microenvironment to reproduce cancer cell signaling and develop drug screens for anti-cancer agents.
- Published
- 2018
- Full Text
- View/download PDF
10. Stem Cells and Cancer Stem Cells
- Author
-
Van Pham, Phuc and Pham, Phuc Van
- Published
- 2015
- Full Text
- View/download PDF
11. YAP-Driven Malignant Reprogramming of Epithelial Stem Cells at Single Cell Resolution.
- Author
-
Gutkind JS, Faraji F, Ramirez S, Clubb L, Sato K, Quiroz PA, Galloway W, Mikulski Z, Hoang T, Medetgul-Ernar K, Marangoni P, Jones K, Officer A, Molinolo A, Kim K, Sakaguchi K, Califano J, Smith Q, Klein O, and Tamayo P
- Abstract
Tumor initiation represents the first step in tumorigenesis during which normal progenitor cells undergo cell fate transition to cancer. Capturing this process as it occurs in vivo , however, remains elusive. Here we employ cell tracing approaches with spatiotemporally controlled oncogene activation and tumor suppressor inhibition to unveil the processes underlying oral epithelial progenitor cell reprogramming into cancer stem cells (CSCs) at single cell resolution. This revealed the rapid emergence of a distinct stem-like cell state, defined by aberrant proliferative, hypoxic, squamous differentiation, and partial epithelial to mesenchymal (pEMT) invasive gene programs. Interestingly, CSCs harbor limited cell autonomous invasive capacity, but instead recruit myeloid cells to remodel the basement membrane and ultimately initiate tumor invasion. CSC transcriptional programs are conserved in human carcinomas and associated with poor patient survival. These findings illuminate the process of cancer initiation at single cell resolution, thus identifying candidate targets for early cancer detection and prevention., Competing Interests: DECLARATION OF INTERESTS J.S.G. has received other commercial research support from Kura Oncology, Mavupharma, Dracen, Verastem, and SpringWorks Therapeutics, and is a consultant/advisory board member for Domain Therapeutics, Pangea Therapeutics, and io9, and founder of Kadima Pharmaceuticals. The remaining authors declare no competing interests. Additional Declarations: Yes there is potential Competing Interest. J.S.G. has received other commercial research support from Kura Oncology, Mavupharma, Dracen, SpringWorks Therapeutics, is a consultant/advisory board member for Oncoceutics Inc., Vividion Therapeutics, Domain Therapeutics, and Pangea Therapeutics, and io9, and founder of Kadima Pharmaceuticals. The remaining authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
12. The pro-tumorigenic effects of metabolic alterations in glioblastoma including brain tumor initiating cells.
- Author
-
Libby, Catherine J., Tran, Anh Nhat, Scott, Sarah E., Griguer, Corinne, and Hjelmeland, Anita B.
- Subjects
- *
CELL metabolism , *GLIOBLASTOMA multiforme , *BRAIN tumors , *CANCER cells , *OXIDATIVE phosphorylation - Abstract
De-regulated cellular energetics is an emerging hallmark of cancer with alterations to glycolysis, oxidative phosphorylation, the pentose phosphate pathway, lipid oxidation and synthesis and amino acid metabolism. Understanding and targeting of metabolic reprogramming in cancers may yield new treatment options, but metabolic heterogeneity and plasticity complicate this strategy. One highly heterogeneous cancer for which current treatments ultimately fail is the deadly brain tumor glioblastoma. Therapeutic resistance, within glioblastoma and other solid tumors, is thought to be linked to subsets of tumor initiating cells, also known as cancer stem cells. Recent profiling of glioblastoma and brain tumor initiating cells reveals changes in metabolism, as compiled here, that may be more broadly applicable. We will summarize the profound role for metabolism in tumor progression and therapeutic resistance and discuss current approaches to target glioma metabolism to improve standard of care. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
13. CBP-mediated Wnt3a/β-catenin signaling promotes cervical oncogenesis initiated by Piwil2
- Author
-
Jing Liang, Haiyan Liang, Zhongjun Dong, Weidong Zhao, Ying Zhou, Keqin Yan, Dingqing Feng, Bin Ling, Huan Yu, and Wenhui Wang
- Subjects
0301 basic medicine ,TFs, transcription factors ,Cancer Research ,Cellular differentiation ,Uterine Cervical Neoplasms ,Mice ,IPP, image-pro plus ,Tumor initiating cell ,0302 clinical medicine ,GSEA, gene set enrichment analysis ,Wnt Signaling Pathway ,Original Research ,SCC, squamous carcinoma of the cervix ,Cervical cancer ,Piwil2 ,pCSC, precancerous stem cell ,biology ,Wnt signaling pathway ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,CREB-Binding Protein ,Immunohistochemistry ,LSIL, low-grade squamous intraepithelial lesion ,TCF, T-cell factor ,Gene Expression Regulation, Neoplastic ,Squamous intraepithelial lesion ,Cell Transformation, Neoplastic ,KLF4 ,030220 oncology & carcinogenesis ,Argonaute Proteins ,Neoplastic Stem Cells ,TIC, tumor initiating cell ,Female ,Disease Susceptibility ,PVDF, polyvinylidene fluoride ,Cell Survival ,CBP, CREB binding protein ,IHC, immunohistochemical ,Antineoplastic Agents ,HPV, human papillomavirus ,DICD, differentiation-induced cell death ,CBP ,Models, Biological ,lcsh:RC254-282 ,Kruppel-Like Factor 4 ,03 medical and health sciences ,SOX2 ,Cell Line, Tumor ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,CREB-binding protein ,HSIL, high-grade squamous intraepithelial lesion ,medicine.disease ,Xenograft Model Antitumor Assays ,CSC, cancer stem cell ,Wnt signaling ,FIGO, the international federation of gynecology and obstetrics ,Disease Models, Animal ,030104 developmental biology ,Drug Resistance, Neoplasm ,Cancer research ,biology.protein ,WNT3A - Abstract
Our previous work demonstrated that Piwil2 reactivated by the human papillomavirus oncoproteins E6 and E7 may reprogram somatic cells into tumor-initiating cells (TICs), which contribute to cervical neoplasia lesions. Maintaining the stemness of TICs is critical for the progression of cervical lesions. Here, we determined that canonical Wnt signaling was aberrantly activated in HaCaT cells transfected with lentivirus expressing Piwil2 and in cervical lesion specimens of low-grade squamous intraepithelial lesion, high-grade squamous intraepithelial lesion, and invasive carcinoma. Blocking the β-catenin and CREB binding protein interaction with ICG-001 significantly downregulated the reprogramming factors c-Myc, Nanog, Oct4, Sox2, and Klf4, thus leading to cell differentiation and preventing tumorigenicity in Piwil2-overexpressing HaCaT cells. Similarly, Piwil2 also critically regulated the canonical Wnt signaling pathway in cervical cancer. We further demonstrated that ICG-001 increased cisplatin sensitivity and significantly suppressed tumor growth of cervical cancer alone or in combination with cisplatin both in vitro and in vivo. The β-catenin/ CREB binding protein-mediated transcription activated by Piwil2 is essential for the maintenance of TICs, therefore contributing to the progression of cervical oncogenesis.
- Published
- 2021
14. Elimination of cancer stem cells and reactivation of latent HIV-1 via AMPK activation: Common mechanism of action linking inhibition of tumorigenesis and the potential eradication of HIV-1.
- Author
-
Finley, Jahahreeh
- Subjects
CANCER stem cells ,EMBRYONIC stem cells ,HIGHLY active antiretroviral therapy ,T cells ,APOPTOSIS ,THERAPEUTICS ,HIV infection complications ,CARCINOGENESIS ,ANIMALS ,CELL differentiation ,HIV infections ,IMMUNITY ,IMMUNOLOGY technique ,MATHEMATICAL models ,PHOSPHOTRANSFERASES ,PROTEIN kinases ,STEM cells ,VIRAL physiology ,THEORY ,NEOPLASTIC cell transformation - Abstract
Although promising treatments are currently in development to slow disease progression and increase patient survival, cancer remains the second leading cause of death in the United States. Cancer treatment modalities commonly include chemoradiation and therapies that target components of aberrantly activated signaling pathways. However, treatment resistance is a common occurrence and recent evidence indicates that the existence of cancer stem cells (CSCs) may underlie the limited efficacy and inability of current treatments to effectuate a cure. CSCs, which are largely resistant to chemoradiation therapy, are a subpopulation of cancer cells that exhibit characteristics similar to embryonic stem cells (ESCs), including self-renewal, multi-lineage differentiation, and the ability to initiate tumorigenesis. Interestingly, intracellular mechanisms that sustain quiescence and promote self-renewal in adult stem cells (ASCs) and CSCs likely also function to maintain latency of HIV-1 in CD4+ memory T cells. Although antiretroviral therapy is highly effective in controlling HIV-1 replication, the persistence of latent but replication-competent proviruses necessitates the development of compounds that are capable of selectively reactivating the latent virus, a method known as the "shock and kill" approach. Homeostatic proliferation in central CD4+ memory T (TCM) cells, a memory T cell subset that exhibits limited self-renewal and differentiation and is a primary reservoir for latent HIV-1, has been shown to reinforce and stabilize the latent reservoir in the absence of T cell activation and differentiation. HIV-1 has also been found to establish durable and long-lasting latency in a recently discovered subset of CD4+ T cells known as T memory stem (TSCM) cells. TSCM cells, compared to TCM cells, exhibit stem cell properties that more closely match those of ESCs and ASCs, including self-renewal and differentiation into all memory T cell subsets. It is our hypothesis that activation of AMPK, a master regulator of cellular metabolism that plays a critical role in T cell activation and differentiation of ESCs and ASCs, will lead to both T cell activation-induced latent HIV-1 reactivation, facilitating virus destruction, as well as "activation", differentiation, and/or apoptosis of CSCs, thus inhibiting tumorigenesis. We also propose the novel observation that compounds that have been shown to both facilitate latent HIV-1 reactivation and promote CSC differentiation/apoptosis (e.g. bryostatin-1, JQ1, metformin, butyrate, etc.) likely do so through a common mechanism of AMPK activation. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
15. Bmi1 Severs as a Potential Tumor-Initiating Cell Marker and Therapeutic Target in Esophageal Squamous Cell Carcinoma
- Author
-
Yong Bao, Jianwen Chen, Hui Han, Liang Peng, Wenjing Liao, Maosheng Cheng, Xiaochen Wang, Kang Li, Demeng Chen, Zhi Chen, Ganping Wang, and Fangfang Chen
- Subjects
Cisplatin ,Article Subject ,biology ,business.industry ,Cell ,Therapeutic effect ,macromolecular substances ,Cell Biology ,biology.organism_classification ,Tumor initiating cell ,RC31-1245 ,Esophageal squamous cell carcinoma ,digestive system diseases ,medicine.anatomical_structure ,BMI1 ,Lineage tracing ,Murine leukemia virus ,medicine ,Cancer research ,business ,Internal medicine ,neoplasms ,Molecular Biology ,Research Article ,medicine.drug - Abstract
Esophageal squamous cell carcinoma (ESCC) is a frequent malignant tumor with low 5-year overall survival. Targeting ESCC tumor-initiating cells (TICs) may provide a new research avenue to achieve better therapeutic effects of ESCC. However, the identity and characteristics of ESCC TICs remain poorly understood. Through genetic lineage tracing approach, we found that a group of Moloney murine leukemia virus insertion site 1- (Bmi1-) expressing cell populations present in the invasive front of the esophageal epithelium, providing a continuous flow of tumor cells for ESCC. Subsequently, we found that ablation of Bmi1+ cells from mice with ESCC led to inhibition of tumor growth. In addition, our results demonstrated that PTC-209, an inhibitor of Bmi1, was able to inhibit ESCC progression when combined with cisplatin. In summary, our data suggest that Bmi1+ cells serve as TICs in ESCC.
- Published
- 2020
16. 331 Tumor markers associated with increased survival in a phase II trial of dendritic cell/tumor-initiating-cell vaccine AV-GBM-1 in patients with newly diagnosed glioblastoma
- Author
-
Renato V. LaRocca, Frank P.K. Hsu, Robert O. Dillman, Christopher Duma, Candace Hsieh, Santosh Kesari, Gabriel Nistor, Mehrdad Abedi, David Piccioni, Thomas H. Taylor, Jose Carrillo, Robert Aiken, Xiao-Tang Kong, and Daniela A. Bota
- Subjects
Pharmacology ,Cancer Research ,business.industry ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Dendritic cell ,Newly diagnosed ,Tumor initiating cell ,medicine.disease ,Oncology ,medicine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,In patient ,business ,RC254-282 ,Glioblastoma - Abstract
BackgroundStandard aggressive therapy of glioblastoma (GBM), which includes maximum safe resection, concurrent radiation therapy and temozolomide chemotherapy (RT/TMZ) followed by maintenance TMZ, is associated with a 25% 2-year overall survival (OS). Adding treatment with AV-GBM-1, a vaccine consisting of autologous dendritic cells (DC) pulsed with autologous tumor antigens (ATA) may improve OS by inducing and/or enhancing the host anti-GBM immune response. Methylation of the O-6-methylguanine-DNA methyltransferase (MGMT) gene promoter, and mutation of the gene for isocitrate dehydrogenase (IDH) are favorable prognostic markers in newly diagnosed GBM. An objective of a multi-center phase II clinical trial was to determine whether these markers were still prognostic for OS in patients treated with adjunctive AV-GBM-1.MethodsKey eligibility criteria for intent-to-treat (ITT) enrollment were: (1) confirmation of primary GBM, (2) successful GBM cell culture, (3) collection of sufficient numbers of monocytes (MC) by leukapheresis, (4) Karnofsky Performance Status 70 or greater after recovery from surgery, and (5) plan to treat with concurrent RT/TMZ. AV-GBM-1 was manufactured while patients were being treated with RT/TMZ. Interleukin-4 and granulocyte-macrophage colony stimulating factor (GM-CSF) were used to differentiate DC from MC. Each vaccine consisted of autologous DC incubated with ATA from the lysate of irradiated cultured GBM cells grown in serum-free media with factors that favor survival and proliferation of stem cells and early progenitor cells (tumor-initiating cells). After recovery from RT/TMZ, intent was to vaccinate for up to six months with cryopreserved AV-GBM-1 admixed with 500 mg GM-CSF. All patients had testing for MGMT-methylation and IDH-mutation. OS was calculated from date of ITT enrollment.Results60 patients were enrolled during August 2018 to January 2020. MGMT promoter methylation was detected in 21 (35%), mutated IDH in 7 (12%), and one or both in 25 (42%). At a minimum follow-up of 15 months, median OS had not been reached for patients with a methylated MGMT promotor, IDH mutation, or one or both, compared to 14.6 months for 38 with unmethylated MGMT promotor (p=0.026), 14.7 months for 53 with IDH wild-type (p=0.044), and 14.6 months for 35 who had neither (p=0.017). 18-month OS rates were 59% vs 35% for MGMT promotor methylation, 71% vs 40% for IDH mutation and 58% vs 32% for either.ConclusionsBoth MGMT promotor methylation and IDH mutation were associated with a substantial and similar survival benefit in primary GBM patients treated with AV-GBM-1 in addition to standard aggressive therapy.Trial RegistrationClinicaltrialsgov NCT03400917Ethics ApprovalThis study was approved by the Western IRB, approval number 20182582; all participants gave written informed consent before taking part
- Published
- 2021
17. WIP Drives Tumor Progression through YAP/TAZ-Dependent Autonomous Cell Growth.
- Author
-
Gargini, Ricardo, Escoll, Maribel, García, Esther, García-Escudero, Ramón, Wandosell, Francisco, and Antón, Inés María
- Abstract
Summary In cancer, the deregulation of growth signaling pathways drives changes in the cell’s architecture and its environment that allow autonomous growth of tumors. These cells then acquire a tumor-initiating “stemness” phenotype responsible for disease advancement to more aggressive stages. Here, we show that high levels of the actin cytoskeleton-associated protein WIP (WASP-interacting protein) correlates with tumor growth, both of which are linked to the tumor-initiating cell phenotype. We find that WIP controls tumor growth by boosting signals that stabilize the YAP/TAZ complex via a mechanism mediated by the endocytic/endosomal system. When WIP levels are high, the β-catenin Adenomatous polyposis coli (APC)-axin-GSK3 destruction complex is sequestered to the multi-vesicular body compartment, where its capacity to degrade YAP/TAZ is inhibited. YAP/TAZ stability is dependent on Rac, p21-activated kinase (PAK) and mammalian diaphanous-related formin (mDia), and is Hippo independent. This close biochemical relationship indicates an oncogenic role for WIP in the physiology of cancer pathology by increasing YAP/TAZ stability. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
18. TGFβ promotes breast cancer stem cell self-renewal through an ILEI/LIFR signaling axis
- Author
-
Toros Dincman, Breege V. Howley, Simon Grelet, Alec N. Woosley, Shaun K. Olsen, Bidyut K. Mohanty, Annamarie C. Dalton, Philip H. Howe, George S. Hussey, and Sean Bloos
- Subjects
0301 basic medicine ,Cancer Research ,Leukemia Inhibitory Factor Receptor alpha Subunit ,Cell ,Leukemia inhibitory factor receptor ,Heterogeneous-Nuclear Ribonucleoproteins ,Metastasis ,tumor initiating cell ,hnRNP E1 ,0302 clinical medicine ,Mice, Inbred NOD ,Transforming Growth Factor beta ,Breast cancer stem cell ,Cell Self Renewal ,RNA-Binding Proteins ,3. Good health ,Neoplasm Proteins ,DNA-Binding Proteins ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cytokines ,Female ,Signal transduction ,Stem cell ,Signal Transduction ,STAT3 Transcription Factor ,Epithelial-Mesenchymal Transition ,Breast Neoplasms ,Biology ,Article ,Fam3C ,03 medical and health sciences ,PCBP1 ,Cancer stem cell ,Cell Line, Tumor ,Genetics ,medicine ,Gene silencing ,metastasis ,Animals ,Humans ,Molecular Biology ,ILEI ,LIFR ,Mammary Neoplasms, Experimental ,Epithelial Cells ,medicine.disease ,030104 developmental biology ,Tumor progression ,Cancer research - Abstract
FAM3C/Interleukin-like EMT Inducer (ILEI) is an oncogenic member of the FAM3 cytokine family and serves essential roles in both epithelial-mesenchymal transition (EMT) and breast cancer metastasis. ILEI expression levels are regulated through a non-canonical TGFβ signaling pathway by 3′-UTR-mediated translational silencing at the mRNA level by hnRNP E1. TGFβ stimulation or silencing of hnRNP E1 increases ILEI translation and induces an EMT program that correlates with enhanced invasion and migration. Recently, EMT has been linked to the formation of breast cancer stem cells (BCSCs) that confer both tumor cell heterogeneity as well as chemoresistant properties. Herein, we demonstrate that hnRNP E1 knockdown significantly shifts normal mammary epithelial cells to mesenchymal BCSCs in vitro and in vivo. We further validate that modulating ILEI protein levels results in the abrogation of these phenotypes, promoting further investigation into the unknown mechanism of ILEI signaling that drives tumor progression. We identify LIFR as the receptor for ILEI, which mediates signaling through STAT3 to drive both EMT and BCSC formation. Reduction of either ILEI or LIFR protein levels results in reduced tumor growth, fewer tumor initiating cells and reduced metastasis within the hnRNP E1 knock-down cell populations in vivo. These results reveal a novel ligand-receptor complex that drives the formation of BCSCs and represents a unique target for the development of metastatic breast cancer therapies.
- Published
- 2019
19. Functional States in Tumor-Initiating Cell Differentiation in Human Colorectal Cancer
- Author
-
Ava Oberlack, Mathias Kalxdorf, Katharina Jechow, Mark Kriegsmann, Stephan M Tirier, Jeroen Krijgsveld, Christian Conrad, Roland Eils, Karin Laaber, Torsten Müller, Teresa G Krieger, Martin Schneider, Friederike Herbst, Mario Huerta, Sebastian M. Dieter, Claudia R. Ball, Robert Lorenz Chua, Foo Wei Ten, Martina K. Zowada, Katharina Kriegsmann, Jeongbin Park, and Hanno Glimm
- Subjects
0301 basic medicine ,Metabolic state ,Cancer Research ,Colorectal cancer ,Cell ,colorectal cancer ,Biology ,tumor-initiating cells ,tumor cell differentiation ,lcsh:RC254-282 ,Article ,Tumor Initiating Cells ,03 medical and health sciences ,0302 clinical medicine ,tumor heterogeneity ,medicine ,single-cell RNA-sequencing ,Treatment resistance ,patient-derived cancer models ,Cell subpopulations ,medicine.disease ,Tumor initiating cell ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,transcriptional programs ,Cancer cell ,Cancer research ,tumor metabolism - Abstract
Simple Summary Different types of cells with tumor-initiating cell (TIC) activity contribute to colorectal cancer (CRC) progression and resistance to anti-cancer treatment. In this study, we aimed to understand whether different cell types exist within a patient-derived tumor culture, distinguishable by different patterns of their gene expression. By mRNA sequencing of patient-derived CRC cultures at the single-cell level, we defined expression programs that closely resemble differentiated cell populations of the normal intestine. Here, cell type-associated subpopulations showed differences in functional properties such as cell growth and energy metabolism. Subsequent functional analyses in vitro and in vivo demonstrated that metabolic states are linked to TIC activity in primary CRC cultures. We also show that TIC activity is dependent on oxidative phosphorylation, which may therefore represent a target for novel therapies. Abstract Intra-tumor heterogeneity of tumor-initiating cell (TIC) activity drives colorectal cancer (CRC) progression and therapy resistance. Here, we used single-cell RNA-sequencing of patient-derived CRC models to decipher distinct cell subpopulations based on their transcriptional profiles. Cell type-specific expression modules of stem-like, transit amplifying-like, and differentiated CRC cells resemble differentiation states of normal intestinal epithelial cells. Strikingly, identified subpopulations differ in proliferative activity and metabolic state. In summary, we here show at single-cell resolution that transcriptional heterogeneity identifies functional states during TIC differentiation. Furthermore, identified expression signatures are linked to patient prognosis. Targeting transcriptional states associated to cancer cell differentiation might unravel novel vulnerabilities in human CRC.
- Published
- 2021
20. Redox biology in normal cells and cancer: Restoring function of the redox/Fyn/c-Cbl pathway in cancer cells offers new approaches to cancer treatment.
- Author
-
Noble, Mark, Mayer-Pröschel, Margot, Li, Zaibo, Dong, Tiefei, Cui, Wanchang, Pröschel, Christoph, Ambeskovic, Ibro, Dietrich, Joerg, Han, Ruolan, Yang, Yin Miranda, Folts, Christopher, Stripay, Jennifer, Chen, Hsing-Yu, and Stevens, Brett M.
- Subjects
- *
CANCER treatment , *CANCER chemotherapy , *AUTOPOIESIS , *CANCER cells , *CELLULAR signal transduction , *CELL division - Abstract
This review discusses a unique discovery path starting with novel findings on redox regulation of precursor cell and signaling pathway function and identification of a new mechanism by which relatively small changes in redox status can control entire signaling networks that regulate self-renewal, differentiation, and survival. The pathway central to this work, the redox/Fyn/c-Cbl (RFC) pathway, converts small increases in oxidative status to pan-activation of the c-Cbl ubiquitin ligase, which controls multiple receptors and other proteins of central importance in precursor cell and cancer cell function. Integration of work on the RFC pathway with attempts to understand how treatment with systemic chemotherapy causes neurological problems led to the discovery that glioblastomas (GBMs) and basal-like breast cancers (BLBCs) inhibit c-Cbl function through altered utilization of the cytoskeletal regulators Cool-1/βpix and Cdc42, respectively. Inhibition of these proteins to restore normal c-Cbl function suppresses cancer cell division, increases sensitivity to chemotherapy, disrupts tumor-initiating cell (TIC) activity in GBMs and BLBCs, controls multiple critical TIC regulators, and also allows targeting of non-TICs. Moreover, these manipulations do not increase chemosensitivity or suppress division of nontransformed cells. Restoration of normal c-Cbl function also allows more effective harnessing of estrogen receptor-α (ERα)-independent activities of tamoxifen to activate the RFC pathway and target ERα-negative cancer cells. Our work thus provides a discovery strategy that reveals mechanisms and therapeutic targets that cannot be deduced by standard genetics analyses, which fail to reveal the metabolic information, isoform shifts, protein activation, protein complexes, and protein degradation critical to our discoveries. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
21. Phosphoprotein analysis reveals MEK inhibition as a way to target non-small cell lung cancer tumor initiating cells.
- Author
-
Lundholm, Lovisa, Hååg, Petra, Juntti, Therese, Lewensohn, Rolf, and Viktorsson, Kristina
- Subjects
- *
PHOSPHOPROTEINS , *LUNG cancer treatment , *PROTEIN-tyrosine kinases , *CANCER cells , *IONIZING radiation , *CANCER chemotherapy - Abstract
Purpose: We aimed to analyze the activation status of commonly deregulated receptor tyrosine kinases (RTK) in human non-small cell lung cancer (NSCLC) tumor initiating cells (TIC) previously demonstrated to be refractory to ionizing radiation and chemotherapy. Materials and methods: Phosphorylated RTK and important signaling nodes were assayed using PathScan RTK Signaling Antibody Array Kit in NSCLC TIC and bulk cells 4 h post-irradiation (IR) and validated by Western blot. The effect of mitogen- activated protein kinase kinase (MEK) inhibition combined with IR was analyzed using clonogenic assay. Results: H125 TIC displayed decreased basal phosphorylation of insulin-like growth factor 1 receptor (IGF-1R) and signal transducer and activator of transcription 1 (STAT1) (Tyr701) as compared to bulk cells. Total IGF-1R levels were significantly lower in NSCLC TIC as compared to bulk cells. A higher degree of extracellular signal-regulated kinase (ERK) phosphorylation was evident in TIC and concordantly MEK inhibition reduced TIC viability. Moreover, MEK inhibition also decreased clonogenicity upon IR suggesting that MEK and downstream signaling impart on TIC radiation response. Conclusions: We demonstrate reduced basal phosphorylation of several signaling pathways including lower total IGF-1R levels in NSCLC TIC which is of potential concern for RTK inhibitor use. Importantly, MEK inhibition decreased cell viability of NSCLC TIC alone or in combination with IR. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
22. Cells of origin in osteosarcoma: Mesenchymal stem cells or osteoblast committed cells?
- Author
-
Mutsaers, Anthony J. and Walkley, Carl R.
- Subjects
- *
OSTEOSARCOMA , *MESENCHYMAL stem cells , *OSTEOBLASTS , *THERAPEUTICS , *BONE cancer , *CANCER diagnosis - Abstract
Abstract: Osteosarcoma is a disease with many complex genetic abnormalities but few well defined genetic drivers of tumor initiation and evolution. The disease is diagnosed and defined through the observation of malignant osteoblastic cells that produce osteoid, however the exact cell of origin for this cancer remains to be definitively defined. Evidence exists to support a mesenchymal stem cell as well as committed osteoblast precursors as the cell of origin. Increasing numbers of experimental models have begun to shed light on to the likely cell population that gives rise to OS in vivo with the weight of evidence favoring an osteoblastic population as the cell of origin. As more information is gathered regarding osteosarcoma initiating cells and how they may relate to the cell of origin we will derive a better understanding of the development of this disease which may ultimately lead to clinical improvements through more personalized therapeutic approaches. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
23. Cool-1-Mediated Inhibition of c-Cbl Modulates Multiple Critical Properties of Glioblastomas, Including the Ability to Generate Tumors In Vivo.
- Author
-
Stevens, Brett M., Folts, Christopher J., Cui, Wanchang, Bardin, Addie L., Walter, Kevin, Carson-Walter, Eleanor, Vescovi, Angelo, and Noble, Mark
- Subjects
GLIOMAS ,CELL proliferation ,TUMORS ,UBIQUITIN ligases ,ANTIGENS ,CARMUSTINE ,ALKYLATING agents - Abstract
We discovered that glioblastoma (GBM) cells use Cool-1/β-pix to inhibit normal activation of the c-Cbl ubiquitin ligase via the redox/Fyn/c-Cbl pathway and that c-Cbl inhibition is critical for GBM cell function. Restoring normal c-Cbl activity by Cool-1 knockdown in vitro reduced GBM cell division, almost eliminated generation of adhesion-independent spheroids, reduced the representation of cells expressing antigens thought to identify tumor initiating cells (TICs), reduced levels of several proteins of critical importance in TIC function (such as Notch-1 and Sox2), and increased sensitivity to BCNU (carmustine) and temozolomide (TMZ). In vivo, Cool-1 knockdown greatly suppressed the ability of GBM cells to generate tumors, an outcome that was c-Cbl dependent. In contrast, Cool-1 knockdown did not reduce division or increase BCNU or TMZ sensitivity in primary glial progenitor cells and Cool-1/c-Cbl complexes were not found in normal brain tissue. Our studies provide the first evidence that Cool-1 may be critical in the biology of human tumors, that suppression of c-Cbl by Cool-1 may be critical for generation of at least a subset of GBMs and offer a novel target that appears to be selectively necessary for TIC function and modulates chemoresistance in GBM cells. Targeting such proteins that inhibit c-Cbl offers potentially attractive opportunities for therapeutic development. S tem C ells 2014;32:1124-1135 [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
24. Partial least squares based gene expression analysis in estrogen receptor positive and negative breast tumors.
- Author
-
W. MA, T.-F . ZHANG, P . LU, and S.H. LU
- Abstract
BACKGROUND: Breast cancer is categorized into two broad groups: estrogen receptor positive (ER+) and ER negative (ER-) groups. Previous study proposed that under trastuzumab-based neoadjuvant chemotherapy, tumor initiating cell (TIC) featured ER- tumors response better than ER+ tumors. Exploration of the molecular difference of these two groups may help developing new therapeutic strategies, especially for ER- patients. MATERIALS AND METHODS:With gene expression profile from the Gene Expression Omnibus (GEO) database, we performed partial least squares (PLS) based analysis, which is more sensitive than common variance/regression analysis. RESULTS: We acquired 512 differentially expressed genes. Four pathways were found to be enriched with differentially expressed genes, involving immune system, metabolism and genetic information processing process. Network analysis identified five hub genes with degrees higher than 10, including APP, ESR1, SMAD3, HDAC2, and PRKAA1. CONCLUSIONS: Our findings provide new understanding for the molecular difference between TIC featured ER- and ER+ breast tumors with the hope offer supports for therapeutic studies. [ABSTRACT FROM AUTHOR]
- Published
- 2014
25. All-trans retinoic acid potentiates the chemotherapeutic effect of cisplatin by inducing differentiation of tumor initiating cells in liver cancer.
- Author
-
Zhang, Yang, Guan, Dong-xian, Shi, Jie, Gao, Hong, Li, Jing-jing, Zhao, Jiang-sha, Qiu, Lin, Liu, Jiang, Li, Nan, Guo, Wei-xing, Xue, Jie, Zhou, Fei-guo, Wu, Meng-chao, Wang, Hong-yang, Xie, Dong, and Cheng, Shu-qun
- Subjects
- *
CANCER cell differentiation , *LIVER cancer , *TRETINOIN , *CANCER chemotherapy , *CISPLATIN , *LIVER cancer patients , *ADJUVANT treatment of cancer - Abstract
Background & Aims: Systemic chemotherapy serves as an adjuvant treatment for post-operation patients with hepatocellular carcinoma (HCC), and provides curative option for the patients with unresectable HCC. However, its efficiency is largely limited because of the high incidence of chemo-resistance. Increasing evidence has shown that tumor initiating cells (TICs) not only have the ability to self-renew and drive the initiation and progression of cancer, but also exhibit greater resistance to conventional chemo- and radio-therapies than non-TICs. It was the aim of this study to investigate the effects of ATRA with and without cisplatin on TIC differentiation and apoptosis in human HCC. Methods: In the present study, we evaluated the TICs of HCC cell differentiation induced by all-trans retinoic acid (ATRA), and developed a novel chemotherapeutic approach to HCC, by characterizing the function of combinatorial treatment with cis-diammineplatinum(II) (cisplatin) and ATRA in vitro and in vivo. Results: ATRA effectively induced differentiation of TICs, which potentiated the cytotoxic effects of cisplatin. The combinatorial treatment of ATRA acid and cisplatin reduced protein kinase B (AKT) (Thr308) phosphorylation, and promoted apoptosis of HCC cells more significantly than treatment with cisplatin alone. In addition, the combined treatment with the two drugs exerted stronger inhibition on either HCC cell migration in vitro or metastasis in vivo, when compared to the treatment with either drug alone. Conclusions: These results indicated that ATRA could significantly improve the effect of cisplatin, which is at least partially attributed to ATRA-induced differentiation of HCC TICs, and the subsequent decrease in this chemo-resistant subpopulation. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
26. 332 Tumor collection and establishment of tumor-initiating cell cultures as antigen source for AV-GBM-1 dendritic cell vaccines for a phase II trial in patients with newly diagnosed glioblastoma
- Author
-
Candace Hsieh, Robert O. Dillman, Frank P.K. Hsu, Gabriel Nistor, David Piccioni, Christopher Duma, Santosh Kesari, Jose Carrillo, Robert Aiken, Thomas H. Taylor, Daniela A. Bota, Xiao-Tang Kong, Renato V. LaRocca, and Mehrdad Abedi
- Subjects
Pharmacology ,Cancer Research ,business.industry ,Immunology ,Dendritic cell ,Newly diagnosed ,Tumor initiating cell ,medicine.disease ,Oncology ,Antigen ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Medicine ,In patient ,business ,Glioblastoma - Abstract
BackgroundDespite standard aggressive therapy (maximum safe surgical resection, concurrent radiation therapy and temozolomide chemotherapy (RT/TMZ), then maintenance TMZ), 2-year survival is only about 25% for patients with newly diagnosed primary glioblastoma (GBM). Adding AV-GBM-1, a vaccine consisting of autologous dendritic cells (DC) pulsed with autologous tumor antigens (ATA) may improve survival. One objective of a multi-center phase II clinical trial was to determine the feasibility of collecting fresh GBM and establishing short-term cell cultures of GBM tumor-initiating cells (TIC) to serve as ATA source.MethodsKey eligibility criteria for tumor collection were (1) clinical suspicion of new primary GBM, (2) age 18 to 70 years (3) tentative agreement to undergo a leukapheresis procedure after recovery from surgery, and (4) tentative plans for RT/TMZ. Fresh tumor was placed in media and shipped in a transport kit by overnight courier to AIVITA where a cell suspension was placed in culture and incubated in serum-free medium with factors that favor survival and proliferation of TICS (stem cells and early progenitor cells). The intent was to produce a patient-specific DC-ATA vaccine by incubating a lysate of irradiated TICs with autologous DC for subsequent subcutaneous injection.ResultsPatients were enrolled from five sites in California, one in Kentucky and one in New Jersey. Tumors were collected between August 2018 and January 2020. 106 patients consented for tumor collection, but 15 were not GBM, 4 had insufficient tissue to send, 2 patients withdrew consent, 4 were ineligible because of age, and 1 was ineligible because of autoimmune disease. Of the 80 GBM tumors that were placed into culture, 7 were discontinued because of patient withdrawal. 71/73 (97%) resulted in a successful cell culture; two were unsuccessful because of contamination. 60/71 subsequently consented for intent-to-treat ; 46/60 (77%) had cells in culture for 28 days or less, 11 were in culture for 30 to 35 days, and the remaining 3 were cultured 46, 54, and 55 days. The average number of cells per culture at the time of irradiation was 14.0 million (range 0.78 to 63.3 million). 58/60 (97%) yielded more than 1 million TICs for irradiation for the tumor cell lysate; 36/60 (60%) had more than 10 million cells irradiated. 57 patients were subsequently treated with AV-GBM-1 after recovery from RT/TMZ.ConclusionsSelf-renewing GBM TIC cultures can be reliably and rapidly established for use as the antigen source for personal DC-ATA vaccines.Trial RegistrationClinicaltrialsgov NCT03400917Ethics ApprovalThis study was approved by the Western IRB, approval number 20182582; all participants gave written informed consent before taking part
- Published
- 2021
27. 333 Changes in proteomic markers after injections of personal AV-GBM-1 dendritic cell/tumor initiating cell vaccines in a phase II trial in patients with newly diagnosed glioblastoma
- Author
-
Thomas H. Taylor, Candace Hsieh, Xiao-Tang Kong, Frank P.K. Hsu, Gabriel Nistor, Christopher Duma, Santosh Kesari, Jose Carrillo, Aleksandra J. Poole, David Piccioni, Daniela A. Bota, Renato V. LaRocca, Mehrdad Abedi, Robert Aiken, and Robert O. Dillman
- Subjects
Pharmacology ,Cancer Research ,business.industry ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Newly diagnosed ,Dendritic cell ,medicine.disease ,Tumor initiating cell ,Oncology ,medicine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,In patient ,business ,RC254-282 ,Glioblastoma - Abstract
BackgroundDespite standard aggressive therapy, including maximum safe surgical resection, concurrent radiation therapy and temozolomide chemotherapy (RT/TMZ) followed by maintenance TMZ, survival is still extremely poor for patients with newly diagnosed primary glioblastoma (GBM). Adding treatment with AV-GBM-1, a personal vaccine consisting of autologous dendritic cells (DC) pulsed with autologous tumor antigens (ATA) may improve survival. One objective of a multi-center phase II clinical trial was to determine changes in blood proteomics before and after injections of AV-GBM-1.MethodsAV-GBM-1 consists of autologous DC incubated with ATA from a lysate of irradiated autologous GBM cells that had been placed in culture and incubated in serum-free medium with factors that favor the survival and proliferation of stem cells and early progenitor cells. After recovery from RT/TMZ, GBM patients were injected subcutaneously with AV-GBM-1 admixed in granulocyte-macrophage colony-stimulating factor (GM-CSF) at weeks 1, 2, 3, 8, 12, 16, 20, and 24. Blood samples obtained at baseline (week-0), just prior to the third injection (week-2) and just prior to the fourth injection (week-8), were cryopreserved and subsequently analyzed for 448 proteomic markers using quantitative, multiplex enzyme-linked immunosorbent assays (Raybiotech, Inc., Norcross, GA.). In this preliminary analysis the averages of paired samples for each time point were determined and compared using the student T-Test with a focus on differences of pResultsPatients were enrolled from five sites in California, and one each in Kentucky and New Jersey. 57 patients were treated during November 2018 to October 2020. Paired samples from all three time points were available for 49 patients. After two weekly injections there were increases in thymus-and activation-regulated chemokine (TARC, CCL17), the chemotactic protein chemerin, lipocalin-2, (expressed by macrophages and epithelium in response to inflammation) and angiopoietin-1 (suppressor of vascular inflammation), and decreases in thrombospondin-5 (possibly involved in synaptogenesis in brain repair), angiotensinogen (a precursor of all angiotensin peptides), and beta-fibroblast growth factor (important in tissue repair). The increase in TARC (pConclusionsIf there were humoral changes in proteins associated with Th1 and Th2 responses, these were no longer present after two weekly vaccinations. More sophisticated analyses of this data set, such as principal component analysis, may be needed to understand the effects of AV-GBM-1.Trial RegistrationClinicaltrialsgov NCT03400917Ethics ApprovalThis study was approved by the Western IRB, approval number 20182582; all participants gave written informed consent before taking part
- Published
- 2021
28. Perspectives on cancer stem cells in osteosarcoma.
- Author
-
Basu-Roy, Upal, Basilico, Claudio, and Mansukhani, Alka
- Subjects
- *
CANCER stem cells , *OSTEOSARCOMA , *CANCER chemotherapy , *BONE cancer treatment , *PROGENITOR cells , *CANCER cell proliferation - Abstract
Abstract: Osteosarcoma is an aggressive pediatric tumor of growing bones that, despite surgery and chemotherapy, is prone to relapse. These mesenchymal tumors are derived from progenitor cells in the osteoblast lineage that have accumulated mutations to escape cell cycle checkpoints leading to excessive proliferation and defects in their ability to differentiate appropriately into mature bone-forming osteoblasts. Like other malignant tumors, osteosarcoma is often heterogeneous, consisting of phenotypically distinct cells with features of different stages of differentiation. The cancer stem cell hypothesis posits that tumors are maintained by stem cells and it is the incomplete eradication of a refractory population of tumor-initiating stem cells that accounts for drug resistance and tumor relapse. In this review we present our current knowledge about the biology of osteosarcoma stem cells from mouse and human tumors, highlighting new insights and unresolved issues in the identification of this elusive population. We focus on factors and pathways that are implicated in maintaining such cells, and differences from paradigms of epithelial cancers. Targeting of the cancer stem cells in osteosarcoma is a promising avenue to explore to develop new therapies for this devastating childhood cancer. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
29. Determining Mammosphere-Forming Potential: Application of the Limiting Dilution Analysis.
- Author
-
Rota, Lauren, Lazzarino, Deborah, Ziegler, Amber, LeRoith, Derek, and Wood, Teresa
- Subjects
- *
EPITHELIAL cells , *MAMMARY gland tumors , *TUMORS , *STEM cells , *CANCER cells , *CELL lines - Abstract
Originally adapted from the neurosphere assay, the nonadherent mammosphere assay has been utilized to assess early progenitor/stem cell frequency in a given population of mammary epithelial cells. This method has also been used to measure the frequency of tumorsphere initiating cells in both primary mammary tumors as well as in tumor cell lines. Although, the mammosphere assay has been used extensively in the mammary gland field, a standard method of quantifying and analyzing sphere growth in this assay has remained undefined. Here, we discuss the use and benefit of using a limiting dilution analysis to quantify sphere-forming frequency in primary mammary epithelial cells grown in nonadherent conditions. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
30. Side population cells in human cancers
- Author
-
Wu, Colleen and Alman, Benjamin A.
- Subjects
- *
CANCER cells , *STEM cells , *MULTIPLE tumors , *DRUG therapy , *DYES & dyeing , *MICROBIOLOGICAL techniques - Abstract
Abstract: Cancer stem cells (CSCs) are found in multiple tumor types. While the presence of surface markers selectively expressed on CSCs are used to isolate these cells, no marker or pattern of makers are known to prospectively identify CSCs in many tumor types. In such cases exploitation of stem cell characteristics can be used to identify CSCs and one such characteristic is the capacity to extrude dyes such as Hoechst 33342. Cell that exclude this dye are referred to as side population (SP) cells. These cells share characteristics of CSCs, specifically, they are enriched for tumor initiating capacity, they express stem-like genes, and they are resistant to chemotherapeutic drugs. Dye exclusion is a valuable technique as it identifies a unique population of cells with stem-like characteristics. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
31. Succession of transiently active tumor‐initiating cell clones in human pancreatic cancer xenografts
- Author
-
Benedikt Brors, Christof von Kalle, Stefan Frohling, Moritz Koch, Christopher M. Hoffmann, Ulrich Abel, Sebastian M. Dieter, Jürgen Weitz, Manfred G. Schmidt, Claudia R. Ball, Wilko Weichert, Felix Oppel, Frank Bergmann, Jens Werner, Claudia Scholl, Taronish D. Dubash, Karl Roland Ehrenberg, Hanno Glimm, Naveed Ishaque, and Friederike Herbst
- Subjects
Male ,clonal dynamics ,0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Pathology ,medicine.medical_specialty ,Tics ,Xenotransplantation ,medicine.medical_treatment ,pancreatic cancer ,Cell ,Population ,Adenocarcinoma ,Biology ,phenotypic plasticity ,03 medical and health sciences ,0302 clinical medicine ,Mice, Inbred NOD ,Pancreatic cancer ,mental disorders ,medicine ,Animals ,Humans ,education ,Research Articles ,Cancer ,education.field_of_study ,Stem Cells ,Metastasis formation ,Tumor initiating cell ,medicine.disease ,nervous system diseases ,Pancreatic Neoplasms ,body regions ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,tumor‐initiating cells ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Cancer research ,Heterografts ,Molecular Medicine ,Female ,Digestive System ,human activities ,Research Article - Abstract
Although tumor‐initiating cell (TIC) self‐renewal has been postulated to be essential in progression and metastasis formation of human pancreatic adenocarcinoma (PDAC), clonal dynamics of TICs within PDAC tumors are yet unknown. Here, we show that long‐term progression of PDAC in serial xenotransplantation is driven by a succession of transiently active TICs producing tumor cells in temporally restricted bursts. Clonal tracking of individual, genetically marked TICs revealed that individual tumors are generated by distinct sets of TICs with very little overlap between subsequent xenograft generations. An unexpected functional and phenotypic plasticity of pancreatic TICs in vivo underlies the recruitment of inactive TIC clones in serial xenografts. The observed clonal succession of TIC activity in serial xenotransplantation is in stark contrast to the continuous activity of limited numbers of self‐renewing TICs within a fixed cellular hierarchy observed in other epithelial cancers and emphasizes the need to target TIC activation, rather than a fixed TIC population, in PDAC.
- Published
- 2017
32. Targeting ADAM17 inhibits human colorectal adenocarcinoma progression and tumor-initiating cell frequency
- Author
-
Theodore H. Welling, Kathryn E. Luker, Robert E. Hollingsworth, Ronald Herbst, Judith S. Sebolt-Leopold, Jonathan Rios-Doria, Eric R. Fearon, Elizabeth Ziemke, Matt Flynn, Joseph Dosch, Suneetha Thomas, Elaine M. Hurt, Karin M. Hardiman, and Shanshan Wan
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Notch ,Colorectal cancer ,Notch signaling pathway ,03 medical and health sciences ,Therapeutic approach ,0302 clinical medicine ,Internal medicine ,medicine ,Colorectal adenocarcinoma ,colorectal ,ADAM17 ,TACE ,Tumor microenvironment ,business.industry ,medicine.disease ,Tumor initiating cell ,3. Good health ,stem cell ,Specific antibody ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunology ,Stem cell ,business ,Research Paper - Abstract
// Joseph Dosch 1, * , Elizabeth Ziemke 1, * , Shanshan Wan 2 , Kathryn Luker 1 , Theodore Welling 2 , Karin Hardiman 2 , Eric Fearon 3 , Suneetha Thomas 4 , Matthew Flynn 4 , Jonathan Rios-Doria 4 , Robert Hollingsworth 4 , Ronald Herbst 4 , Elaine Hurt 4, # and Judith Sebolt-Leopold 1, # 1 Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA 2 Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA 3 Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA 4 Department of Oncology Research, MedImmune, LLC, Gaithersburg, MD 20878, USA * These authors contributed equally to this work # Co-senior authors Correspondence to: Judith Sebolt-Leopold, email: jssl@med.umich.edu Keywords: ADAM17, TACE, colorectal, stem cell, Notch Received: November 24, 2016 Accepted: April 03, 2017 Published: May 10, 2017 ABSTRACT ADAM17 (a disintegrin and metalloproteinase 17)/TACE (TNFα converting enzyme) has emerged as a potential therapeutic target in colorectal cancer (CRC) and other cancers, due in part to its role in regulating various tumor cell surface proteins and growth factors and cytokines in the tumor microenvironment. The emergence of MEDI3622, a highly potent and specific antibody-based ADAM17 inhibitor, has allowed testing of the concept that targeting ADAM17 may be an important new therapeutic approach for CRC patients. We demonstrate that MEDI3622 is highly efficacious on tumor growth in multiple human CRC PDX models, resulting in improved survival of animals bearing tumor xenografts. MEDI3622 was further found to impact Notch pathway activity and tumor-initiating cells. The promising preclinical activity seen here supports further clinical investigation of this treatment approach to improve therapeutic outcome for patients diagnosed with metastatic CRC, including patients with KRAS-mutant tumors for whom other therapeutic options are currently limited.
- Published
- 2017
33. Hypoxia-induced Autophagy Drives Colorectal Cancer Initiation and Progression by Activating the PRKC/PKC-EZR (Ezrin) Pathway
- Author
-
Martin Nurmik, Daniel Val, Komal Qureshi-Baig, Sonia Frasquilho, Pit Ullmann, Serge Haan, Bassam Janji, Diana Kuhn, Michel Mittelbronn, Petr V. Nazarov, Elodie Viry, Eric Koncina, Nikolaus Zuegel, Yervand Karapetyan, Laurent Antunes, Martine Schmitz, Fabien Rodriguez, Elisabeth Letellier, Marc Boulmont, Vitaly I. Pozdeev, and ELEVIE (28504270, R-AGR-3140), ondation Cancer (grant F1R-LSC-PAU-13HY2C), Fonds National de la Recherche (FNR) (under the AFR grant scheme and PRIDE scheme) [sponsor]
- Subjects
0301 basic medicine ,Carcinogenesis ,Colorectal cancer ,Mice, SCID ,Biochemistry, biophysics & molecular biology [F05] [Life sciences] ,Autophagy-Related Protein 5 ,tumor-initiating cell ,Ezrin ,Mice, Inbred NOD ,Cell Self Renewal ,Phosphorylation ,Hypoxia ,Biochimie, biophysique & biologie moléculaire [F05] [Sciences du vivant] ,Protein Kinase C ,Tumor initiating cell ,Phenotype ,Disease Progression ,Neoplastic Stem Cells ,medicine.symptom ,Colorectal Neoplasms ,Signal Transduction ,Research Article ,Research Paper ,congenital, hereditary, and neonatal diseases and abnormalities ,cancer stem cell ,Colon ,colorectal cancer ,self-renewal capacity ,Biology ,03 medical and health sciences ,Cancer stem cell ,mental disorders ,medicine ,Autophagy ,Animals ,Humans ,Molecular Biology ,Protein kinase C ,030102 biochemistry & molecular biology ,hypoxia ,Autophagosomes ,Cell Biology ,Hypoxia (medical) ,medicine.disease ,nervous system diseases ,ezrin ,body regions ,Cytoskeletal Proteins ,030104 developmental biology ,Cancer research ,human activities ,protein kinase C - Abstract
In solid tumors, cancer stem cells (CSCs) or tumor-initiating cells (TICs) are often found in hypoxic niches. Nevertheless, the influence of hypoxia on TICs is poorly understood. Using previously established, TIC-enrichedpatient-derived colorectal cancer (CRC) cultures, we show that hypoxia increases the self-renewal capacity of TICs while inducing proliferation arrest in their more differentiated counterpart cultures. Gene expression data revealed macroautophagy/autophagy as one of the major pathways induced by hypoxia in TICs. Interestingly, hypoxia-induced autophagy was found to induce phosphorylation of EZR (ezrin) at Thr567 residue, which could be reversed by knocking down ATG5, BNIP3, BNIP3L, or BECN1. Furthermore, we identified PRKCA/PKCα as a potential kinase involved in hypoxia-induced autophagy-mediated TIC self-renewal. Genetic targeting of autophagy or pharmacological inhibition of PRKC/PKC and EZR resulted in decreased tumor-initiating potential of TICs. In addition, we observed significantly reduced in vivo tumor initiation and growth after a stable knockdown of ATG5. Analysis of human CRC samples showed that p-EZR is often present in TICs located in the hypoxic and autophagic regions of the tumor. Altogether, our results establish the hypoxia-autophagy-PKC-EZR signaling axis as a novel regulatory mechanism of TIC self-renewal and CRC progression. Autophagy inhibition might thus represent a promising therapeutic strategy for cancer patients. Abbreviations ATG: autophagy related; BECN1: beclin 1; BNIP3: BCL2 interacting protein 3; BNIP3L: BCL2 interacting protein 3 like; CQ: chloroquine; CSC: cancer stem cells; CRC: colorectal cancer; HIF1A/HIF-1α: hypoxia inducible factor 1 subunit alpha; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; PRKC/PKC: protein kinase C; SQSTM1/p62: sequestosome 1; TICs: tumor-initiating cells.
- Published
- 2019
34. Corrigendum to 'NRF2/SHH signaling cascade promotes tumor-initiating cell lineage and drug resistance in hepatocellular carcinoma' [Canc. Lett. 476 (2020) 48–56]
- Author
-
Shao Hang Cai, Etienne Ho Kit Mok, Hua Jian Yu, Carmen Oi Ning Leung, Hoi Wing Leung, Martina Mang Leng Lei, Victor W.S. Ma, Jing Ping Yun, William C. Cho, Terence Kin Wah Lee, Stephanie Ma, Irene Oi-Lin Ng, and Eunice Yuen Ting Lau
- Subjects
Cancer Research ,Lineage (genetic) ,Oncology ,Shh signaling ,Hepatocellular carcinoma ,Cancer research ,medicine ,Drug resistance ,Biology ,Tumor initiating cell ,medicine.disease - Published
- 2021
35. WIP Drives Tumor Progression through YAP/TAZ-Dependent Autonomous Cell Growth
- Author
-
Maribel Escoll, Inés M. Antón, Francisco Wandosell, Esther García, Ricardo Gargini, Ramón García-Escudero, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad (España), and European Commission
- Subjects
0301 basic medicine ,Hippo pathway ,β-catenin destruction complex ,Endocytic cycle ,Cell ,Polymerization ,tumor initiating cell ,actin dynamics ,Neoplasms ,glioma ,WIPF1 ,beta Catenin ,Protein Stability ,Kinase ,Intracellular Signaling Peptides and Proteins ,Multivesicular Bodies ,Endocytosis ,Cell biology ,Phenotype ,medicine.anatomical_structure ,Disease Progression ,Neoplastic Stem Cells ,Signal transduction ,Signal Transduction ,Proteasome Endopeptidase Complex ,Epithelial-Mesenchymal Transition ,Cell Survival ,Adenomatous polyposis coli ,proliferation ,Endosomes ,macromolecular substances ,Biology ,multivesicular body ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,breast cancer ,Cell Line, Tumor ,medicine ,Humans ,YAP/TAZ ,Neoplasm Invasiveness ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Hippo signaling pathway ,Cell growth ,YAP-Signaling Proteins ,Phosphoproteins ,Actins ,Cytoskeletal Proteins ,030104 developmental biology ,Tumor progression ,Transcriptional Coactivator with PDZ-Binding Motif Proteins ,Proteolysis ,Trans-Activators ,biology.protein ,Transcription Factors - Abstract
In cancer, the deregulation of growth signaling pathways drives changes in the cell¿s architecture and its environment that allow autonomous growth of tumors. These cells then acquire a tumor-initiating ¿stemness¿ phenotype responsible for disease advancement to more aggressive stages. Here, we show that high levels of the actin cytoskeleton-associated protein WIP (WASP-interacting protein) correlates with tumor growth, both of which are linked to the tumor-initiating cell phenotype. We find that WIP controls tumor growth by boosting signals that stabilize the YAP/TAZ complex via a mechanism mediated by the endocytic/endosomal system. When WIP levels are high, the ß-catenin Adenomatous polyposis coli (APC)-axin-GSK3 destruction complex is sequestered to the multi-vesicular body compartment, where its capacity to degrade YAP/TAZ is inhibited. YAP/TAZ stability is dependent on Rac, p21-activated kinase (PAK) and mammalian diaphanous-related formin (mDia), and is Hippo independent. This close biochemical relationship indicates an oncogenic role for WIP in the physiology of cancer pathology by increasing YAP/TAZ stability., MINECO (SAF2013-45937-R to I.M.A.) and MINECO/FEDER (SAF2015-70368-R to I.M.A. and F.W.), the European Union (EU-FP7-2009-CT222887 to F.W.), the Instituto de Salud Carlos III Centro de Investigación Biomédica en Red (CIBERNED to F.W.) and by a grant from ISCIII-RETIC (RD12/0036/0009 to R.G.E.)
- Published
- 2016
36. Piwil2 is reactivated by HPV oncoproteins and initiates cell reprogrammingviaepigenetic regulation during cervical cancer tumorigenesis
- Author
-
Ying Zhou, Dingqing Feng, Bin Ling, Weidong Zhao, Keqin Yan, Jing Liang, Haiyan Liang, and Zhongjun Dong
- Subjects
0301 basic medicine ,Homeobox protein NANOG ,Carcinogenesis ,cervical cancer ,Papillomavirus E7 Proteins ,Mice, Nude ,Uterine Cervical Neoplasms ,HPV oncoprotein ,Cell Growth Processes ,Epigenetic Repression ,medicine.disease_cause ,Malignant transformation ,tumor initiating cell ,Kruppel-Like Factor 4 ,Mice ,03 medical and health sciences ,SOX2 ,Cell Line, Tumor ,Animals ,Humans ,Medicine ,RNA, Small Interfering ,Piwil2 ,Human papillomavirus 16 ,business.industry ,cell reprogramming ,Cancer ,Oncogene Proteins, Viral ,Cellular Reprogramming ,medicine.disease ,Xenograft Model Antitumor Assays ,Embryonic stem cell ,Gene Expression Regulation, Neoplastic ,Repressor Proteins ,030104 developmental biology ,Oncology ,KLF4 ,Argonaute Proteins ,Immunology ,Neoplastic Stem Cells ,Cancer research ,Female ,Transcriptome ,business ,Reprogramming ,Priority Research Paper - Abstract
// Dingqing Feng 1 , Keqin Yan 1 , Ying Zhou 2 , Haiyan Liang 1 , Jing Liang 1 , Weidong Zhao 2,3 , Zhongjun Dong 4 and Bin Ling 1 1 Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, China 2 Department of Obstetrics and Gynecology, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei, China 3 Department of Gynecology and Oncology, Anhui Provincial Cancer Hospital, Hefei, China 4 School of Medicine, Tsinghua University, Beijing, China Correspondence to: Bin Ling, email: // Keywords : Piwil2, cell reprogramming, tumor initiating cell, HPV oncoprotein, cervical cancer Received : April 04, 2016 Accepted : July 19, 2016 Published : September 01, 2016 Abstract The human papillomavirus (HPV) oncoproteins E6 and E7 are risk factors that are primarily responsible for the initiation and progression of cervical cancer, and they play a key role in immortalization and transformation by reprogramming differentiating host epithelial cells. It is unclear how cervical epithelial cells transform into tumor-initiating cells (TICs). Here, we observed that the germ stem cell protein Piwil2 is expressed in pre-cancerous and malignant lesions of the cervix and cervical cancer cell lines with the exception of the non-HPV-infected C33a cell line. Knockdown of Piwil2 by shRNA led to a marked reduction in proliferation and colony formation, in vivo tumorigenicity, chemo-resistance, and the proportion of cancer stem-like cells. In contrast, Piwil2 overexpression induced malignant transformation of HaCaT cells and the acquisition of tumor-initiating capabilities. Gene-set enrichment analysis revealed embryonic stem cell (ESC) identity, malignant biological behavior, and specifically, activation targets of the cell reprogramming factors c-Myc , Klf4 , Nanog , Oct4 , and Sox2 in Piwil2 -overexpressing HaCaT cells. We further confirmed that E6 and E7 reactivated Piwil2 and that E6 and E7 overexpression resulted in a similar gene-set enrichment pattern as Piwil2 overexpression in HaCaT cells. Moreover, Piwil2 overexpression or E6 and E7 activation induced H3K9 acetylation but reduced H3K9 trimethylation, which contributed to the epigenetic reprogramming and ESC signature maintenance, as predicted previously. Our study demonstrates that Piwil2 , reactivated by the HPV oncoproteins E6 and E7, plays an essential role in the transformation of cervical epithelial cells to TICs via epigenetics-based cell reprogramming.
- Published
- 2016
37. Tumor-initiating cell frequency is relevant for glioblastoma aggressiveness
- Author
-
Daniela Osti, Massimiliano Del Bene, Bianca Pollo, Cristina Richichi, Giuliana Pelicci, Francesco DiMeco, Monica Patanè, and Lorenzo Fornasari
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,congenital, hereditary, and neonatal diseases and abnormalities ,Pathology ,medicine.medical_specialty ,Tumor incidence ,Tics ,Neuropathology ,Mice ,03 medical and health sciences ,neurosphere ,Limiting dilution ,Internal medicine ,Neurosphere ,mental disorders ,medicine ,Animals ,Humans ,Progression-free survival ,Aged ,Aged, 80 and over ,Brain Neoplasms ,business.industry ,glioblastoma ,Middle Aged ,Tumor initiating cell ,medicine.disease ,tumor-initiating cell frequency ,nervous system diseases ,body regions ,limiting dilution assay ,030104 developmental biology ,Neoplastic Stem Cells ,tumorigenicity ,Female ,business ,human activities ,Research Paper ,Glioblastoma - Abstract
// Cristina Richichi 1 , Daniela Osti 1 , Massimiliano Del Bene 2 , Lorenzo Fornasari 1 , Monica Patane 3 , Bianca Pollo 3 , Francesco DiMeco 2, 4 , Giuliana Pelicci 1, 5 1 Department of Experimental Oncology, European Institute of Oncology, 20139, Milan, Italy 2 Department of Neurosurgery, IRCCS Foundation Neurological Institute “C. Besta”, 20133, Milan, Italy 3 Department of Neuropathology, IRCCS Foundation Neurological Institute “C. Besta”, 20133, Milan, Italy 4 Department of Neurosurgery, Johns Hopkins University, Baltimore, MD 21218, USA 5 Department of Translational Medicine, Piemonte Orientale University “Amedeo Avogadro”, 28100 Novara, Italy Correspondence to: Giuliana Pelicci, email: giuliana.pelicci@ieo.eu Keywords: glioblastoma, tumor-initiating cell frequency, neurosphere, limiting dilution assay, tumorigenicity Received: March 03, 2016 Accepted: August 13, 2016 Published: August 25, 2016 ABSTRACT Glioblastoma (GBM) is maintained by a small subpopulation of tumor-initiating cells (TICs). The arduous assessment of TIC frequencies challenges the prognostic role of TICs in predicting the clinical outcome in GBM patients. We estimated the TIC frequency in human GBM injecting intracerebrally in mice dissociated cells without any passage in culture. All GBMs contained rare TICsand were tumorigenic in vivo but only 54% of them grew in vitro as neurospheres. We demonstrated that neurosphere formation in vitro did not foretell tumorigenic ability in vivo and frequencies calculated in vitro overestimated the TIC content. Our findings assert the pathological significance of GBM TICs. TIC number correlated positively with tumor incidence and inversely with survival of tumor-bearing mice. Stratification of GBM patients according to TIC content revealed that patients with low TIC frequency experienced a trend towards a longer progression free survival. The expression of either putative stem-cell markers or markers associated with different GBM molecular subtypes did not associate with either TIC content or neurosphere formation underlying the limitations of TIC identification based on the expression of some putative stem cell-markers.
- Published
- 2016
38. Kinomic profiling of glioblastoma cells reveals PLCG1 as a target in restricted glucose
- Author
-
Walker, Kiera, Boyd, Nathaniel H., Anderson, Joshua C., Willey, Christopher D., and Hjelmeland, Anita B.
- Published
- 2018
- Full Text
- View/download PDF
39. Kinomic profiling of glioblastoma cells reveals PLCG1 as a target in restricted glucose
- Author
-
Christopher D. Willey, Joshua C. Anderson, Nathaniel H. Boyd, Anita B. Hjelmeland, and Kiera Walker
- Subjects
0301 basic medicine ,Clinical Biochemistry ,Kinomics ,Tumor initiating cell ,PLCG1 ,03 medical and health sciences ,Protein phosphorylation ,Kinase activity ,Tumor microenvironment ,Phospholipase C ,Chemistry ,Kinase ,Research ,Cancer stem cell ,lcsh:RM1-950 ,Biochemistry (medical) ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,Cancer cell ,Cancer research ,Molecular Medicine ,Phosphorylation ,Restricted glucose ,Glioblastoma - Abstract
Background For glioblastoma (GBM) treatments to be effective in vivo, understanding the effects of the tumor microenvironment is imperative. In traditional cell culture conditions, glucose concentrations do not model physiologic levels, nor the diminished concentrations found in tumor niches. We therefore sought to profile the differences in kinase activity in GBM cells cultured in restricted glucose to identify pathways that could be targeted with small molecule inhibitors. Methods Using the PamStation12 platform, we examined the ability of GBM lysates from cells cultured in standard or low glucose conditions to phosphorylate 144 tyrosine and 144 serine/threonine peptides that correspond to known protein phosphorylation sites. Potential kinase targets were identified and validated using small molecule kinase inhibitors in GBM spheroid cultures. Results Using results from two GBM patient-derived xenografts, we determined common changes to peptides derived from Phospholipase C, Gamma 1 (PLCG1) and Raf-1. Using PLC and Raf inhibitors, we found a significantly stronger growth inhibitory effect of the PLC inhibitor U73122 under restricted glucose conditions. In contrast, Raf inhibitors were significantly growth inhibitory regardless of the nutrient level tested. Conclusions Together, our data demonstrate that kinase activity is altered in low glucose conditions and that kinomic profiling can assist with the identification of effective strategies to target GBM growth. Our data further suggest the importance of accurately modeling the tumor microenvironment to reproduce cancer cell signaling and develop drug screens for anti-cancer agents.
- Published
- 2018
40. Alternative Treatments For Melanoma: Targeting BCL-2 Family Members to De-Bulk and Kill Cancer Stem Cells
- Author
-
David A. Norris, Nabanita Mukherjee, Mayumi Fujita, Yiqun G. Shellman, and Josianna V. Schwan
- Subjects
Complementary Therapies ,Male ,Oncology ,medicine.medical_specialty ,Skin Neoplasms ,Antineoplastic Agents ,Apoptosis ,Dermatology ,Risk Assessment ,Tumor heterogeneity ,Biochemistry ,Article ,Cancer stem cell ,Internal medicine ,Humans ,Medicine ,Molecular Targeted Therapy ,Melanoma ,neoplasms ,Molecular Biology ,Survival analysis ,business.industry ,Treatment regimen ,Bcl-2 family ,Cancer ,Cell Biology ,Prognosis ,Tumor initiating cell ,medicine.disease ,Survival Analysis ,3. Good health ,Treatment Outcome ,Proto-Oncogene Proteins c-bcl-2 ,Drug Resistance, Neoplasm ,Immunology ,Neoplastic Stem Cells ,Female ,Neoplasm Recurrence, Local ,business - Abstract
For the first time new treatments in melanoma have produced significant responses in advanced diseases, but 30–90% of melanoma patients do not respond or eventually relapse after the initial response to the current treatments. The resistance of these melanomas is likely due to tumor heterogeneity, which may be explained by models such as the stochastic, hierarchical, and phenotype-switching models. This review will discuss the recent advancements in targeting BCL-2 family members for cancer treatments, and how this approach can be applied as an alternative option to combat melanoma, and overcome melanoma relapse or resistance in current treatment regimens.
- Published
- 2015
- Full Text
- View/download PDF
41. CDC20 maintains tumor initiating cells
- Author
-
William A. Flavahan, Leo J.Y. Kim, Qiulian Wu, Stephen C. Mack, Jeremy N. Rich, Chengwei Chu, Shideng Bao, Christopher G. Hubert, Kailin Yang, and Qi Xie
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,congenital, hereditary, and neonatal diseases and abnormalities ,Chromatin Immunoprecipitation ,cancer stem cell ,Cdc20 Proteins ,Cell Survival ,Blotting, Western ,Population ,CDC20 ,tumor initiating cell ,Mice ,Mice, Inbred NOD ,Cancer stem cell ,glioma ,Glioma ,mental disorders ,Survivin ,Tumor Cells, Cultured ,Animals ,Humans ,Medicine ,education ,Cell Proliferation ,education.field_of_study ,Brain Neoplasms ,business.industry ,Forkhead Box Protein M1 ,glioblastoma ,Forkhead Transcription Factors ,medicine.disease ,Molecular medicine ,Neural stem cell ,nervous system diseases ,3. Good health ,body regions ,Oncology ,Immunology ,Neoplastic Stem Cells ,FOXM1 ,Cancer research ,Heterografts ,business ,human activities ,Research Paper - Abstract
// Qi Xie 1 , Qiulian Wu 1 , Stephen C. Mack 1 , Kailin Yang 1, 2 , Leo Kim 1 , Christopher G. Hubert 1 , William A. Flavahan 1, 3, 4, 5 , Chengwei Chu 1, 6 , Shideng Bao 1, 2 , Jeremy N. Rich 1, 2 1 Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA 2 Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA 3 Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA 4 Broad Institute of Harvard and Massachusetts Institute of Technology (MIT), Cambridge, MA 02142, USA 5 Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA 6 Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan Correspondence to: Jeremy N. Rich, e-mail: drjeremyrich@gmail.com Keywords: cancer stem cell, glioblastoma, glioma, tumor initiating cell, CDC20 Received: March 05, 2015 Accepted: April 16, 2015 Published: April 28, 2015 ABSTRACT Glioblastoma is the most prevalent and lethal primary intrinsic brain tumor. Glioblastoma displays hierarchical arrangement with a population of self-renewing and tumorigenic glioma tumor initiating cells (TICs), or cancer stem cells. While non-neoplastic neural stem cells are generally quiescent, glioblastoma TICs are often proliferative with mitotic control offering a potential point of fragility. Here, we interrogate the role of cell-division cycle protein 20 (CDC20), an essential activator of anaphase-promoting complex (APC) E3 ubiquitination ligase, in the maintenance of TICs. By chromatin analysis and immunoblotting, CDC20 was preferentially expressed in TICs relative to matched non-TICs. Targeting CDC20 expression by RNA interference attenuated TIC proliferation, self-renewal and in vivo tumor growth. CDC20 disruption mediated its effects through induction of apoptosis and inhibition of cell cycle progression. CDC20 maintains TICs through degradation of p21 CIP1/WAF1 , a critical negative regulator of TICs. Inhibiting CDC20 stabilized p21 CIP1/WAF1 , resulting in repression of several genes critical to tumor growth and survival, including CDC25C, c-Myc and Survivin. Transcriptional control of CDC20 is mediated by FOXM1, a central transcription factor in TICs. These results suggest CDC20 is a critical regulator of TIC proliferation and survival, linking two key TIC nodes – FOXM1 and p21 CIP1/WAF1 — elucidating a potential point for therapeutic intervention.
- Published
- 2015
42. Reactive oxygen species produced by altered tumor metabolism impacts cancer stem cell maintenance.
- Author
-
Tuy K, Rickenbacker L, and Hjelmeland AB
- Subjects
- Humans, Oxidative Stress, Reactive Oxygen Species, Tumor Microenvironment, Neoplasms, Neoplastic Stem Cells
- Abstract
Controlling reactive oxygen species (ROS) at sustainable levels can drive multiple facets of tumor biology, including within the cancer stem cell (CSC) population. Tight regulation of ROS is one key component in CSCs that drives disease recurrence, cell signaling, and therapeutic resistance. While ROS are well-appreciated to need oxygen and are a product of oxidative phosphorylation, there are also important roles for ROS under hypoxia. As hypoxia promotes and sustains major stemness pathways, further consideration of ROS impacts on CSCs in the tumor microenvironment is important. Furthermore, glycolytic shifts that occur in cancer and may be promoted by hypoxia are associated with multiple mechanisms to mitigate oxidative stress. This altered metabolism provides survival advantages that sustain malignant features, such as proliferation and self-renewal, while producing the necessary antioxidants that reduce damage from oxidative stress. Finally, disease recurrence is believed to be attributed to therapy resistant CSCs which can be quiescent and have changes in redox status. Effective DNA damage response pathways and/or a slow-cycling state can protect CSCs from the genomic catastrophe induced by irradiation and genotoxic agents. This review will explore the delicate, yet complex, relationship between ROS and its pleiotropic role in modulating the CSC., (Copyright © 2021 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
43. Targeting the seeds of small cell lung cancer.
- Author
-
Firestein R., Mahara S., Firestein R., and Mahara S.
- Abstract
The concept of antibody drug conjugates (ADCs), which includes the delivery of cytotoxic drugs to antigen-expressing tumor cells by harnessing the antigen-selectivity of a monoclonal antibody, has the potential to redefine the landscape of translational medicine. With the advent of patient derived xenograft (PDX) models and sophisticated genomic technologies, the identification of a selective antigen can be accurately validated within the appropriate tumor milieu. However, a major biological hurdle in cancer translational medicine is the inherent tumoral heterogeneity, underscoring the importance of targeting the 'right' sub-population of cancer cells. Herein, we review a seminal work highlighting the ability to target a key 'stem-like' cancer sub-population called tumor initiating cells (TICs) using engineered ADCs. While the promise of this approach needs to be validated in the clinical setting, TIC-targeted ADCs offer great hope for circumventing current limitations with conventional ADC therapy.Copyright © Annals of Translational Medicine.
- Published
- 2017
44. STEM-35. GTP CYCLOHYDROLASE I IN TUMOR INITIATING CELL MAINTENANCE AND GLIOBLASTOMA GROWTH: FUNCTIONS AND MECHANISMS
- Author
-
Jennifer S. Pollock, Kiera Walker, Randee Sedaka, Wei Chen, James A. Mobley, Anita B. Hjelmeland, David G. Harrison, Yancey Gillespie, Lauren Hocevar, Darley-Usmar, Sara J. Cooper, James R. Hackney, and Anh Nhat Tran
- Subjects
Cancer Research ,Abstracts ,Oncology ,GTP cyclohydrolase I ,Cancer research ,biology.protein ,medicine ,Neurology (clinical) ,Biology ,medicine.disease ,Tumor initiating cell ,Glioblastoma - Abstract
Glioblastoma (GBM), or grade IV astrocytoma, is a deadly disease due in part to the high degree of intratumoral heterogeneity that contributes to treatment failures. One cellular subset enriched for the capacity to propagate tumor growth and promote therapeutic resistance is brain tumor initiating cells (BTICs). The aim of the study was to define the roles of GTP cyclohydrolase 1 (GCH1) in regulation of BTIC phenotypes via reactive species. We examined GCH1 mRNA and protein expression in patient-derived xenografts, clinical samples and glioma gene expression datasets. GCH1 levels were modulated using lentiviral expression systems, and effects on cell growth, self-renewal, reactive species production and in vitro animal survival were determined. We found that GCH1 RNA and protein expression were increased in BTICs in comparison to non-BTICs, but that GCH1 was not exclusive to the BTIC fraction. Indeed, GCH1 was elevated in GBM in comparison to normal brain. Overexpression of GCH1 in GBM cells increased cell growth in vitro and decreased survival in an intracranial GBM mouse model. In contrast, GCH1 knockdown with short hairpin RNA in GBM cells led to growth inhibition in vitro as well as increased survival in animal models. Mechanistically, GCH1 increased CD44 expression and was critical for controlling reactive species balance, including suppressing reactive oxygen species production. In silico analyses demonstrated that higher GCH1 levels in glioma patients correlate with higher glioma grade, recurrence and worse survival. Together, our data suggest that upregulation of GCH1 in BTICs promotes tumor maintenance and is a key regulator of reactive oxygen species in GBM.
- Published
- 2017
45. Lung cancer tumorigenicity and drug resistance are maintained through ALDHhiCD44hi tumor initiating cells
- Author
-
SK Wong, Junwen Wang, Ka Yan Ho, Jing Liu, Zhi-Jie Xiao, Maria Pik Wong, Vicky Pui-Chi Tin, and Mai Har Sham
- Subjects
Male ,Lung Neoplasms ,medicine.medical_treatment ,Cell ,Aldehyde dehydrogenase ,Mice, SCID ,Disease-Free Survival ,Targeted therapy ,tumor initiating cell ,Mice ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,medicine ,Animals ,Humans ,CD44 ,Lung cancer ,drug resistance ,biology ,Cell cycle ,Aldehyde Dehydrogenase ,medicine.disease ,lung cancer ,medicine.anatomical_structure ,Hyaluronan Receptors ,Oncology ,Apoptosis ,Drug Resistance, Neoplasm ,Gene Knockdown Techniques ,biology.protein ,Cancer research ,Neoplastic Stem Cells ,Heterografts ,Stem cell ,Research Paper - Abstract
// Jing Liu 1 , Zhijie Xiao 1 , Sunny Kit-Man Wong 1 , Vicky Pui-Chi Tin 1 , Ka-Yan Ho 1 , Junwen Wang 2 , Mai-Har Sham 2 , Maria Pik Wong 1 1 Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR China 2 Department of Biochemistry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR China Correspondence: Maria Pik Wong, email: // Keywords : lung cancer, tumor initiating cell, aldehyde dehydrogenase, CD44, drug resistance Received : August 5, 2013 Accepted : August 28, 2013 Published : August 30, 2013 Abstract Limited improvement in long term survival of lung cancer patients has been achieved by conventional chemotherapy or targeted therapy. To explore the potentials of tumor initiating cells (TIC)-directed therapy, it is essential to identify the cell targets and understand their maintenance mechanisms. We have analyzed the performance of ALDH/CD44 co-expression as TIC markers and treatment targets of lung cancer using well-validated in vitro and in vivo analyses in multiple established and patient-derived lung cancer cells. The ALDH hi CD44 hi subset showed the highest enhancement of stem cell phenotypic properties compared to ALDH hi CD44 lo , ALDH lo CD44 hi , ALDH lo CD44 lo cells and unsorted controls. They showed higher invasion capacities, pluripotency genes and epithelial-mesenchymal transition transcription factors expression, lower intercellular adhesion protein expression and higher G2/M phase cell cycle fraction. In immunosuppressed mice, the ALDH hi CD44 hi xenografts showed the highest tumor induction frequency, serial transplantability, shortest latency, largest volume and highest growth rates. Inhibition of sonic Hedgehog and Notch developmental pathways reduced ALDH + CD44 + compartment. Chemotherapy and targeted therapy resulted in higher ALDH hi CD44 hi subset viability and ALDH lo CD44 lo subset apoptosis fraction. ALDH inhibition and CD44 knockdown led to reduced stemness gene expression and sensitization to drug treatment. In accordance, clinical lung cancers containing a higher abundance of ALDH and CD44-coexpressing cells was associated with lower recurrence-free survival. Together, results suggested the ALDH hi CD44 hi compartment was the cellular mediator of tumorigenicity and drug resistance. Further investigation of the regulatory mechanisms underlying ALDH hi CD44 hi TIC maintenance would be beneficial for the development of long term lung cancer control.
- Published
- 2013
46. A Tumor initiating cell-enriched prognostic signature for HER2+:ERα− breast cancer; rationale, new features, controversies and future directions
- Author
-
Sean E. Egan, Jeff C. Liu, and Eldad Zacksenhaus
- Subjects
Receptor, ErbB-2 ,Cell ,Breast Neoplasms ,Cell Growth Processes ,HER2 breast cancer ,Biology ,Mouse models ,Bioinformatics ,Somatic evolution in cancer ,Transcriptome ,Mice ,Tumor initiating cell ,Breast cancer ,Cancer stem cell ,medicine ,Animals ,Humans ,PI3K/AKT/mTOR pathway ,Cancer stem cells ,Prognostic signature ,Cancer ,Prognosis ,medicine.disease ,medicine.anatomical_structure ,Receptors, Estrogen ,Oncology ,Research Perspective ,Neoplastic Stem Cells ,Cancer research ,Female ,Tumor necrosis factor alpha ,Signal Transduction - Abstract
// Jeff C. Liu 1,* , Sean E. Egan 2 and Eldad Zacksenhaus 1,* 1 Division of Cell & Molecular Biology, Toronto General Research Institute - University Health Network, Toronto, Canada 2 Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto Correspondence: Jeff Liu, email: // Eldad Zacksenhaus, email: // Keywords : HER2 breast cancer; Prognostic signature; Tumor initiating cell; Cancer stem cells; Mouse models Received : July 10, 2013 Accepted : July 24, 2013 Published : July 26, 2013 Abstract The high intra- and inter-tumor heterogeneity of many types of cancers, including breast cancer (BC), poses great challenge to development of subtype-specific prognosis. In BC, the classification of tumors as either ERα + (Luminal A and Luminal B), HER2 + (ERα + or ERα - ) or triple-negative (TNBC)(Basal-like, claudin-low) guides both prognostication and therapy. Indeed, prognostic signatures for ERα + BC are being incorporated into clinical use. However, these signatures distinguish between luminal A (low risk) and Luminal B (high risk) BC; signatures that identify low/high risk patients with luminal B BC are yet to be developed. Likewise, no signature is in clinical use for HER2 + or TNBC. The major obstacles to development of robust signatures stem from diversity of BC, clonal evolution and heterogeneity within each subtype. We have recently generated a prognostic signature for HER2 + :ERα - BC based on the identification of genes that were differentially expressed in a tumor-initiating cell (TIC)-enriched fraction versus non-TIC fraction from a mouse model of HER2 + BC (MMTV-Hers/Neu). Here we describe the rationale behind development of this prognosticator, and present new features of the signature, including elevated PI3K pathway activity and low TNFalpha and IFNgamma signaling in high-risk tumors. In addition, we address controversies in the field such as whether random gene expression signatures significantly associate with cancer outcome. Finally, we suggest a guideline for development of prognostic signatures and discuss future directions.
- Published
- 2013
47. Restoring mir122 in human stem-like hepatocarcinoma cells, prompts tumor dormancy through smad-independent TGF-β pathway
- Author
-
Jordi Bruix, Loreto Boix, A.C. Rhodes, J.M. López-Oliva, and Universitat de Barcelona
- Subjects
0301 basic medicine ,miR122 ,Oncologia ,Cellular differentiation ,Cell ,Liver Stem Cell ,Smad Proteins ,SMAD ,Stem cells ,tumor initiating cell ,Mice ,0302 clinical medicine ,Transforming Growth Factor beta ,Càncer ,Cancer ,Liver Neoplasms ,hepatocellular carcinoma ,medicine.anatomical_structure ,Oncology ,KLF4 ,030220 oncology & carcinogenesis ,Disease Progression ,Neoplastic Stem Cells ,Liver cancer ,Cèl·lules mare ,Signal Transduction ,Research Paper ,medicine.medical_specialty ,cancer stem cell ,tumor dormancy ,Carcinoma, Hepatocellular ,Biology ,Càncer de fetge ,03 medical and health sciences ,Kruppel-Like Factor 4 ,Cancer stem cell ,Internal medicine ,Cell Line, Tumor ,Spheroids, Cellular ,medicine ,Animals ,Humans ,Cell Proliferation ,Hepatology ,medicine.disease ,MicroRNAs ,030104 developmental biology ,Endocrinology ,Cell culture ,FOXM1 ,Cancer research ,Dormancy ,Proto-Oncogene Proteins c-akt - Abstract
// Loreto Boix 1 , Juan Manuel Lopez-Oliva 1 , Ana Carolina Rhodes 1 , Jordi Bruix 1 1 Barcelona Clinic Liver Cancer (BCLC) Group, Liver Unit, Hospital Clinic of Barcelona, University of Barcelona, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Fundacio Clinic per a la Recerca Biomedica (FCRB), CIBERehd, 08036 Barcelona, Spain Correspondence to: Loreto Boix, email: lboix@clinic.ub.es Keywords: hepatocellular carcinoma, miR122, cancer stem cell, tumor initiating cell, tumor dormancy Received: February 05, 2016 Accepted: June 29, 2016 Published: September 07, 2016 ABSTRACT miR122 is the prevalent miRNA in adult healthy liver and it is responsible for liver stem cell differentiation towards hepatocyte lineage. Its expression is frequently lost in hepatocellular carcinoma (HCC). We studied the effects of restoring miR122 expression in a distinctive cell line derived from human HCC-BCLC9 cells-with a solid stem-like cell profile, high tumor initiating ability and undetectable miR122 expression. We generated a stable BCLC9 cell line that expresses miR122 (BCLC9-miR122). Restitution of miR122 in BCLC9 cells, decreases cell proliferation rate and reduces significantly tumor size in vivo . BCLC9-miR122 cells down-regulate expression of MYC, KLF4, FOXM1, AKT2 and AKT3 genes and up-regulate FOXO1 and FOXO3A gene expression. In addition, miR122 transfected cells decreased AKT2 kinase activation while decreased FOXO1 and FOXO3A protein inactivation. Reduction in tumor size in BCLC9-miR122 associated with an increase in p38MAPK protein expression and activation leading to a low phospho-ERK1/2 to phospho-p38 ratio. Treatment of miR122 positive cells with an inhibitor of TGFBR1 activation, abolished tumor dormancy program and recovered cell proliferation rate through a Smad-independent TGF-β response. HCC stem-like cells can be directed towards cell differentiation and tumor dormancy by restoring miR122 expression. We demonstrate, for the first time, that dormancy program is achieved through a Smad-independent TGF-β pathway. Reestablishing miR122 expression is a promising therapeutic strategy that would work concurrently reducing tumor aggressiveness and decreasing disease recurrence.
- Published
- 2016
48. Determining Mammosphere-Forming Potential: Application of the Limiting Dilution Analysis
- Author
-
Deborah A. Lazzarino, Teresa L. Wood, Amber N. Ziegler, Lauren M. Rota, and Derek LeRoith
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Tumorsphere ,Mammary gland ,Population ,Cell Culture Techniques ,Breast Neoplasms ,Biology ,Article ,Epithelium ,Tumor initiating cell ,Mammary Glands, Animal ,Cancer stem cell ,Cell Line, Tumor ,Neurosphere ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Breast ,Progenitor cell ,Mammary Glands, Human ,education ,Tumor Stem Cell Assay ,Mammary ,education.field_of_study ,Stem cell ,Cell biology ,medicine.anatomical_structure ,Cellular Microenvironment ,Oncology ,Cell culture ,Neoplastic Stem Cells ,Female ,Mammosphere - Abstract
Originally adapted from the neurosphere assay, the nonadherent mammosphere assay has been utilized to assess early progenitor/stem cell frequency in a given population of mammary epithelial cells. This method has also been used to measure the frequency of tumorsphere initiating cells in both primary mammary tumors as well as in tumor cell lines. Although, the mammosphere assay has been used extensively in the mammary gland field, a standard method of quantifying and analyzing sphere growth in this assay has remained undefined. Here, we discuss the use and benefit of using a limiting dilution analysis to quantify sphere-forming frequency in primary mammary epithelial cells grown in nonadherent conditions.
- Published
- 2012
49. The miR-106b-25 cluster targets Smad7, activates TGF-β signaling, and induces EMT and tumor initiating cell characteristics downstream of Six1 in human breast cancer
- Author
-
Anna L Smith, Douglas S. Micalizzi, Aik Choon Tan, David J. Drasin, Rebecca L. Vartuli, Ritsuko Iwanaga, and Heide L. Ford
- Subjects
TGF-β ,Six1 ,Cancer Research ,Epithelial-Mesenchymal Transition ,Receptor, Transforming Growth Factor-beta Type I ,Regulator ,Breast Neoplasms ,Context (language use) ,Protein Serine-Threonine Kinases ,Biology ,medicine.disease_cause ,Article ,Smad7 Protein ,tumor initiating cell ,03 medical and health sciences ,0302 clinical medicine ,Transforming Growth Factor beta ,Cell Line, Tumor ,microRNA ,Genetics ,medicine ,Humans ,Epithelial–mesenchymal transition ,Molecular Biology ,miRNA ,030304 developmental biology ,Homeodomain Proteins ,0303 health sciences ,R-SMAD ,Transforming growth factor beta ,Up-Regulation ,3. Good health ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,Cell Transformation, Neoplastic ,030220 oncology & carcinogenesis ,Immunology ,MCF-7 Cells ,Cancer research ,biology.protein ,Female ,epithelial-to-mesenchymal transition ,Signal transduction ,Carcinogenesis ,Receptors, Transforming Growth Factor beta ,Signal Transduction - Abstract
The role of TGF-β signaling in tumorigenesis is paradoxical: it can be tumor suppressive or tumor promotional, depending on context. The metastatic regulator, Six1, was recently shown to mediate this switch, providing a novel means to explain this elusive “TGF-β paradox”. Herein, we identify a mechanism by which Six1 activates the tumor promotional arm of TGF-β signaling, via its ability to upregulate the miR-106b-25 microRNA cluster, and further identify a novel function for this cluster of microRNAs. While expression of the miR-106b-25 cluster is known to overcome TGF-β-mediated growth suppression via targeting p21 and BIM, we demonstrate for the first time that this same cluster can additionally target the inhibitory Smad7 protein, resulting in increased levels of the TGF-β type I receptor (TβRI) and downstream activation of TGF-β signaling. We further show that the miR-106b-25 cluster is sufficient to induce an epithelial to mesenchymal transition and a tumor initiating cell phenotype, and that it is required downstream of Six1 to induce these phenotypes. Finally, we demonstrate a significant correlation between miR-106b, Six1, and activated TGF-β signaling in human breast cancers, and further show that high levels of miR-106b and miR-93 in breast tumors significantly predicts shortened time to relapse. These findings expand the spectrum of oncogenic functions of miR-106b-25, and may provide a novel molecular explanation, through the Six1 regulated miR-106b-25 cluster, by which TGF-β signaling shifts from tumor suppressive to tumor promoting.
- Published
- 2012
50. Sox2 maintains self-renewal of tumor initiating cells in osteosarcomas
- Author
-
Timothy B. Rapp, Upal Basu-Roy, Alka Mansukhani, Lalitha Ramanathapuram, Eunjeong Seo, Claudio Basilico, Jennifer A. Perry, and Stuart H. Orkin
- Subjects
musculoskeletal diseases ,Cancer Research ,cancer stem cell ,Sox2 ,Bone Neoplasms ,Biology ,Stem cell marker ,Article ,tumor initiating cell ,03 medical and health sciences ,Mice ,0302 clinical medicine ,SOX2 ,Cancer stem cell ,Cell Line, Tumor ,Cell Self Renewal ,Genetics ,Animals ,Antigens, Ly ,Humans ,mesenchymal tumors ,Progenitor cell ,Molecular Biology ,Wnt Signaling Pathway ,030304 developmental biology ,Cell Proliferation ,0303 health sciences ,Osteosarcoma ,SOXB1 Transcription Factors ,Mesenchymal stem cell ,fungi ,Wnt signaling pathway ,Membrane Proteins ,Cell Differentiation ,differentiation ,Wnt signaling ,3. Good health ,Cell Transformation, Neoplastic ,sarcosphere ,030220 oncology & carcinogenesis ,Immunology ,embryonic structures ,Antigens, Surface ,Cancer research ,Neoplastic Stem Cells ,Stem cell ,Signal Transduction - Abstract
Tumors are thought to be sustained by a reservoir of self-renewing cells, termed tumor-initiating cells or cancer stem cells. Osteosarcomas are high-grade sarcomas derived from osteoblast progenitor cells and are the most common pediatric bone malignancy. In this report we show that the stem cell transcription factor Sox2 is highly expressed in human and murine osteosarcoma (mOS) cell lines as well as in the tumor samples. Osteosarcoma cells have increased ability to grow in suspension as osteospheres, that are greatly enriched in expression of Sox2 and the stem cell marker, Sca-1. Depletion of Sox2 by short-hairpin RNAs in independent mOS-derived cells drastically reduces their transformed properties in vitro and their ability to form tumors. Sox2-depleted osteosarcoma cells can no longer form osteospheres and differentiate into mature osteoblasts. Concomitantly, they exhibit decreased Sca-1 expression and upregulation of the Wnt signaling pathway. Thus, despite other mutations, these cells maintain a requirement for Sox2 for tumorigenicity. Our data indicate that Sox2 is required for osteosarcoma cell self renewal, and that Sox2 antagonizes the pro-differentiation Wnt pathway that can in turn reduce Sox2 expression. These studies define Sox2 as a survival factor and a novel biomarker of self renewal in osteosarcomas, and support a tumor suppressive role for the Wnt pathway in tumors of mesenchymal origin. Our findings could provide the basis for novel therapeutic strategies based on inhibiting Sox2 or enhancing Wnt signaling for the treatment of osteosarcomas.
- Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.