1
|
Soleimani A, Saeedi N, Al-Asady AM, Nazari E, Hanaie R, Khazaei M, Ghorbani E, Akbarzade H, Ryzhikov M, Avan A, Mehr SMH. Colorectal Cancer Stem Cell Biomarkers: Biological Traits and Prognostic Insights. Curr Pharm Des 2024; 30:1386-1397. [PMID: 38623972 DOI: 10.2174/0113816128291321240329050945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 03/08/2024] [Accepted: 03/13/2024] [Indexed: 04/17/2024]
Abstract
Due to self-renewal, differentiation, and limitless proliferation properties, Cancer Stem Cells (CSCs) increase the probability of tumor development. These cells are identified by using CSC markers, which are highly expressed proteins on the cell surface of CSCs. Recently, the therapeutic application of CSCs as novel biomarkers improved both the prognosis and diagnosis outcome of colorectal Cancer. In the present review, we focused on a specific panel of colorectal CSC markers, including LGR5, ALDH, CD166, CD133, and CD44, which offers a targeted and comprehensive analysis of their functions. The selection criteria for these markers cancer were based on their established significance in Colorectal Cancer (CRC) pathogenesis and clinical outcomes, providing novel insights into the CSC biology of CRC. Through this approach, we aim to elevate understanding and stimulate further research for developing effective diagnostic and therapeutic strategies in CRC.
Collapse
Affiliation(s)
- Atena Soleimani
- Department of Biochemistry, Mashhad University of Medical Sciences, Razavi Khorasan, Mashhad, Iran
| | - Nikoo Saeedi
- Medical School, Islamic Azad University, Mashhad, Iran
| | | | - Elnaz Nazari
- Department of Physiology, Mashhad University of Medical Sciences, Razavi Khorasan, Mashhad, Iran
| | - Reyhane Hanaie
- Department of Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Razavi Khorasan, Mashhad, Iran
| | - Majid Khazaei
- Department of Physiology, Mashhad University of Medical Sciences, Razavi Khorasan, Mashhad, Iran
| | - Elnaz Ghorbani
- Department of Microbiology, Mashhad University of Medical Sciences, Razavi Khorasan, Mashhad, Iran
| | - Hamed Akbarzade
- Department of Biochemistry, Mashhad University of Medical Sciences, Razavi Khorasan, Mashhad, Iran
| | - Mikhail Ryzhikov
- Department of Biochemistry, Saint Louis University, St. Louis, MO 63103, USA
| | - Amir Avan
- Department of Genetics, Mashhad University of Medical Sciences, Razavi Khorasan, Mashhad, Iran
| | | |
Collapse
|
2
|
Davalos V, Lovell CD, Von Itter R, Dolgalev I, Agrawal P, Baptiste G, Kahler DJ, Sokolova E, Moran S, Piqué L, Vega-Saenz de Miera E, Fontanals-Cirera B, Karz A, Tsirigos A, Yun C, Darvishian F, Etchevers HC, Osman I, Esteller M, Schober M, Hernando E. An epigenetic switch controls an alternative NR2F2 isoform that unleashes a metastatic program in melanoma. Nat Commun 2023; 14:1867. [PMID: 37015919 PMCID: PMC10073109 DOI: 10.1038/s41467-023-36967-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 02/24/2023] [Indexed: 04/06/2023] Open
Abstract
Metastatic melanoma develops once transformed melanocytic cells begin to de-differentiate into migratory and invasive melanoma cells with neural crest cell (NCC)-like and epithelial-to-mesenchymal transition (EMT)-like features. However, it is still unclear how transformed melanocytes assume a metastatic melanoma cell state. Here, we define DNA methylation changes that accompany metastatic progression in melanoma patients and discover Nuclear Receptor Subfamily 2 Group F, Member 2 - isoform 2 (NR2F2-Iso2) as an epigenetically regulated metastasis driver. NR2F2-Iso2 is transcribed from an alternative transcriptional start site (TSS) and it is truncated at the N-terminal end which encodes the NR2F2 DNA-binding domain. We find that NR2F2-Iso2 expression is turned off by DNA methylation when NCCs differentiate into melanocytes. Conversely, this process is reversed during metastatic melanoma progression, when NR2F2-Iso2 becomes increasingly hypomethylated and re-expressed. Our functional and molecular studies suggest that NR2F2-Iso2 drives metastatic melanoma progression by modulating the activity of full-length NR2F2 (Isoform 1) over EMT- and NCC-associated target genes. Our findings indicate that DNA methylation changes play a crucial role during metastatic melanoma progression, and their control of NR2F2 activity allows transformed melanocytes to acquire NCC-like and EMT-like features. This epigenetically regulated transcriptional plasticity facilitates cell state transitions and metastatic spread.
Collapse
Affiliation(s)
- Veronica Davalos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA.
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain.
| | - Claudia D Lovell
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
| | - Richard Von Itter
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
| | - Igor Dolgalev
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Praveen Agrawal
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine/ Montefiore, Bronx, NY, 10461, USA
| | - Gillian Baptiste
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
| | - David J Kahler
- High Throughput Biology Core, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Elena Sokolova
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
| | - Sebastian Moran
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain
| | - Laia Piqué
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain
| | - Eleazar Vega-Saenz de Miera
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
- The Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Barbara Fontanals-Cirera
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
| | - Alcida Karz
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
| | - Aristotelis Tsirigos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Chi Yun
- High Throughput Biology Core, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Farbod Darvishian
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
| | | | - Iman Osman
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
- The Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Manel Esteller
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Catalonia, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
- Centro de Investigacion Biomedica en Red, Cancer (CIBERONC), Madrid, Spain
| | - Markus Schober
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA.
- The Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
- Department of Cell Biology, New York Grossman University School of Medicine, New York, NY, 10016, USA.
| | - Eva Hernando
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
3
|
Deng R, Zheng H, Cai H, Li M, Shi Y, Ding S. Effects of helicobacter pylori on tumor microenvironment and immunotherapy responses. Front Immunol 2022; 13:923477. [PMID: 35967444 PMCID: PMC9371381 DOI: 10.3389/fimmu.2022.923477] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/04/2022] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori is closely associated with gastric cancer. During persistent infection, Helicobacter pylori can form a microenvironment in gastric mucosa which facilitates the survival and colony formation of Helicobacter pylori. Tumor stromal cells are involved in this process, including tumor-associated macrophages, mesenchymal stem cells, cancer-associated fibroblasts, and myeloid-derived suppressor cells, and so on. The immune checkpoints are also regulated by Helicobacter pylori infection. Helicobacter pylori virulence factors can also act as immunogens or adjuvants to elicit or enhance immune responses, indicating their potential applications in vaccine development and tumor immunotherapy. This review highlights the effects of Helicobacter pylori on the immune microenvironment and its potential roles in tumor immunotherapy responses.
Collapse
Affiliation(s)
- Ruiyi Deng
- Peking University Third Hospital, Research Center of Clinical Epidemiology, Beijing, China
- Peking University Health Science Center, Peking University First Medical School, Beijing, China
| | - Huiling Zheng
- Peking University Third Hospital, Department of Gastroenterology, Beijing, China
| | - Hongzhen Cai
- Peking University Third Hospital, Research Center of Clinical Epidemiology, Beijing, China
- Peking University Health Science Center, Peking University First Medical School, Beijing, China
| | - Man Li
- Peking University Third Hospital, Research Center of Clinical Epidemiology, Beijing, China
- Peking University Health Science Center, Peking University Third Medical School, Beijing, China
| | - Yanyan Shi
- Peking University Third Hospital, Research Center of Clinical Epidemiology, Beijing, China
| | - Shigang Ding
- Peking University Third Hospital, Department of Gastroenterology, Beijing, China
| |
Collapse
|
4
|
Gorczyca G, Wartalski K, Wiater J, Samiec M, Tabarowski Z, Duda M. Anabolic Steroids-Driven Regulation of Porcine Ovarian Putative Stem Cells Favors the Onset of Their Neoplastic Transformation. Int J Mol Sci 2021; 22:ijms222111800. [PMID: 34769230 PMCID: PMC8583785 DOI: 10.3390/ijms222111800] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/27/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022] Open
Abstract
Nandrolone (Ndn) and boldenone (Bdn), the synthetic testosterone analogues with strong anabolic effects, despite being recognized as potentially carcinogenic compounds, are commonly abused by athletes and bodybuilders, which includes women, worldwide. This study tested the hypothesis that different doses of Ndn and Bdn can initiate neoplastic transformation of porcine ovarian putative stem cells (poPSCs). Immunomagnetically isolated poPSCs were expanded ex vivo in the presence of Ndn or Bdn, for 7 and 14 days. Results show that pharmacological doses of both Ndn and Bdn, already after 7 days of poPSCs culture, caused a significant increase of selected, stemness-related markers of cancer cells: CD44 and CD133. Notably, Ndn also negatively affected poPSCs growth not only by suppressing their proliferation and mitochondrial respiration but also by inducing apoptosis. This observation shows, for the first time, that chronic exposure to Ndn or Bdn represents a precondition that might enhance risk of poPSCs neoplastic transformation. These studies carried out to accomplish detailed molecular characterization of the ex vivo expanded poPSCs and their potentially cancerous derivatives (PCDs) might be helpful to determine their suitability as nuclear donor cells (NDCs) for further investigations focused on cloning by somatic cell nuclear transfer (SCNT). Such investigations might also be indispensable to estimate the capabilities of nuclear genomes inherited from poPSCs and their PCDs to be epigenetically reprogrammed (dedifferentiated) in cloned pig embryos generated by SCNT. This might open up new possibilities for biomedical research aimed at more comprehensively recognizing genetic and epigenetic mechanisms underlying not only tumorigenesis but also reversal/retardation of pro-tumorigenic intracellular events.
Collapse
Affiliation(s)
- Gabriela Gorczyca
- Department of Endocrinology, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa 9 Street, 30-387 Krakow, Poland;
| | - Kamil Wartalski
- Department of Histology, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7 Street, 31-034 Krakow, Poland; (K.W.); (J.W.)
| | - Jerzy Wiater
- Department of Histology, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7 Street, 31-034 Krakow, Poland; (K.W.); (J.W.)
| | - Marcin Samiec
- Department of Reproductive Biotechnology and Cryoconservation, National Research Institute of Animal Production, Krakowska 1 Street, 32-083 Balice near Kraków, Poland
- Correspondence: (M.S.); (M.D.)
| | - Zbigniew Tabarowski
- Department of Experimental Hematology, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa 9 Street, 30-387 Krakow, Poland;
| | - Małgorzata Duda
- Department of Endocrinology, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa 9 Street, 30-387 Krakow, Poland;
- Correspondence: (M.S.); (M.D.)
| |
Collapse
|
5
|
Castro‐Gil MP, Sánchez‐Rodríguez R, Torres‐Mena JE, López‐Torres CD, Quintanar‐Jurado V, Gabiño‐López NB, Villa‐Treviño S, del‐Pozo‐Jauner L, Arellanes‐Robledo J, Pérez‐Carreón JI. Enrichment of progenitor cells by 2-acetylaminofluorene accelerates liver carcinogenesis induced by diethylnitrosamine in vivo. Mol Carcinog 2021; 60:377-390. [PMID: 33765333 PMCID: PMC8251613 DOI: 10.1002/mc.23298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/24/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023]
Abstract
The potential role of hepatocytes versus hepatic progenitor cells (HPC) on the onset and pathogenesis of hepatocellular carcinoma (HCC) has not been fully clarified. Because the administration of 2-acetylaminofluorene (2AAF) followed by a partial hepatectomy, selectively induces the HPC proliferation, we investigated the effects of chronic 2AAF administration on the HCC development caused by the chronic administration of the carcinogen diethylnitrosamine (DEN) for 16 weeks in the rat. DEN + 2AAF protocol impeded weight gain of animals but promoted prominent hepatomegaly and exacerbated liver alterations compared to DEN protocol alone. The tumor areas detected by γ-glutamyl transferase, prostaglandin reductase-1, and glutathione S-transferase Pi-1 liver cancer markers increased up to 80% as early as 12 weeks of treatment, meaning 6 weeks earlier than DEN alone. This protocol also increased the number of Ki67-positive cells and those of CD90 and CK19, two well-known progenitor cell markers. Interestingly, microarray analysis revealed that DEN + 2AAF protocol differentially modified the global gene expression signature and induced the differential expression of 30 genes identified as HPC markers as early as 6 weeks of treatment. In conclusion, 2AAF induces the early appearance of HPC markers and as a result, accelerates the hepatocarcinogenesis induced by DEN in the rat. Thus, since 2AAF simultaneously administrated with DEN enriches HPC during hepatocarcinogenesis, we propose that DEN + 2AAF protocol might be a useful tool to investigate the cellular origin of HCC with progenitor features.
Collapse
Affiliation(s)
| | - Ricardo Sánchez‐Rodríguez
- Foundation Istituto di Ricerca Pediatrica‐Città della SperanzaPadovaItaly
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | | | | | | | | | - Saúl Villa‐Treviño
- Department of Cell BiologyCenter for Research and Advanced Studies of the National Polytechnic InstituteCiudad de MéxicoMexico
| | | | - Jaime Arellanes‐Robledo
- Laboratory of Liver DiseasesNational Institute of Genomic MedicineCiudad de MéxicoMexico
- Directorate of CátedrasNational Council of Science and TechnologyCiudad de MéxicoMexico
| | | |
Collapse
|
6
|
Noman ASM, Parag RR, Rashid MI, Islam S, Rahman MZ, Chowdhury AA, Sultana A, Jerin C, Siddiqua A, Rahman L, Nayeem J, Akther S, Baidya S, Shil RK, Rahman M, Shirin A, Mahmud R, Hossain SMI, Sumi SA, Chowdhury A, Basher SB, Hasan A, Bithy S, Aklima J, Chowdhury N, Hasan MN, Banu T, Chowdhury S, Hossain MM, Yeger H, Farhat WA, Islam SS. Chemotherapeutic resistance of head and neck squamous cell carcinoma is mediated by EpCAM induction driven by IL-6/p62 associated Nrf2-antioxidant pathway activation. Cell Death Dis 2020; 11:663. [PMID: 32814771 PMCID: PMC7438524 DOI: 10.1038/s41419-020-02907-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 12/24/2022]
Abstract
Overexpression of epithelial cell adhesion molecule (EpCAM) has been associated with chemotherapeutic resistance, leads to aggressive tumor behavior, and results in an adverse clinical outcome. The molecular mechanism by which EpCAM enrichment is linked to therapeutic resistance via Nrf2, a key regulator of antioxidant genes is unknown. We have investigated the link between EpCAM and the Nrf2 pathway in light of therapeutic resistance using head and neck squamous cell carcinoma (HNSCC) patient tumor samples and cell lines. We report that EpCAM was highly expressed in Nrf2-positive and HPV-negative HNSCC cells. In addition, cisplatin-resistant tumor cells consisted of a higher proportion of EpCAMhigh cells compared to the cisplatin sensitive counterpart. EpCAMhigh populations exhibited resistance to cisplatin, a higher efficiency in colony formation, sphere growth and invasion capacity, and demonstrated reduced reactive oxygen species (ROS) activity. Furthermore, Nrf2 expression was significantly higher in EpCAMhigh populations. Mechanistically, expression of Nrf2 and its target genes were most prominently observed in EpCAMhigh populations. Silencing of EpCAM expression resulted in the attenuation of expressions of Nrf2 and SOD1 concomitant with a reduction of Sox2 expression. On the other hand, silencing of Nrf2 expression rendered EpCAMhigh populations sensitive to cisplatin treatment accompanied by the inhibition of colony formation, sphere formation, and invasion efficiency and increased ROS activity. The molecular mechanistic link between EpCAM expression and activation of Nrf2 was found to be a concerted interaction of interleukin-6 (IL-6) and p62. Silencing of p62 expression in EpCAMhigh populations resulted in the attenuation of Nrf2 pathway activation suggesting that Nrf2 pathway activation promoted resistance to cisplatin in EpCAMhigh populations. We propose that therapeutic targeting the Nrf2-EpCAM axis might be an excellent approach to modulate stress resistance and thereby survival of HNSCC patients enriched in EpCAMhigh populations.
Collapse
Affiliation(s)
- Abu Shadat M Noman
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh.,Department of Pathology, McGill University, Montreal, QC, Canada
| | - Rashed R Parag
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Muhammad I Rashid
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Shafiqul Islam
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Mohammad Z Rahman
- Department of Pathology, Chittagong Medical College Hospital, Chittagong, Bangladesh
| | - Ali A Chowdhury
- Department of Radiotherapy, Chittagong Medical College Hospital, Chittagong, Bangladesh
| | - Afrin Sultana
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Chandsultana Jerin
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Ayesha Siddiqua
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Lutfur Rahman
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Junayed Nayeem
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Sonam Akther
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Sunanda Baidya
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Rajib K Shil
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Mizanur Rahman
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh.,Department of Biochemistry, Rangamati Medical College, Rangamati, Bangladesh
| | - Afsana Shirin
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Reaz Mahmud
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - S M Ikram Hossain
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Sharmin A Sumi
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Arfina Chowdhury
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Shabnam B Basher
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Abul Hasan
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Shammy Bithy
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Jannatul Aklima
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Nabila Chowdhury
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Muhammad N Hasan
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Tahmina Banu
- Chittagong Research Institute for Children Surgery (CRICS), Chittagong, Bangladesh
| | - Srikanta Chowdhury
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Muhammad M Hossain
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Herman Yeger
- Developmental and Stem Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Walid A Farhat
- Division of Pediatric Urology, American Family Children's Hospital, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Syed S Islam
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia. .,School of Medicine, Al-Faisal University, Riyadh, Saudi Arabia.
| |
Collapse
|
7
|
Filip S, Vymetalkova V, Petera J, Vodickova L, Kubecek O, John S, Cecka F, Krupova M, Manethova M, Cervena K, Vodicka P. Distant Metastasis in Colorectal Cancer Patients-Do We Have New Predicting Clinicopathological and Molecular Biomarkers? A Comprehensive Review. Int J Mol Sci 2020; 21:E5255. [PMID: 32722130 PMCID: PMC7432613 DOI: 10.3390/ijms21155255] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) remains a serious health problem worldwide. Approximately half of patients will develop distant metastasis after CRC resection, usually with very poor prognosis afterwards. Because patient performance after distant metastasis surgery remains very heterogeneous, ranging from death within 2 years to a long-term cure, there is a clinical need for a precise risk stratification of patients to aid pre- and post-operative decisions. Furthermore, around 20% of identified CRC cases are at IV stage disease, known as a metastatic CRC (mCRC). In this review, we overview possible molecular and clinicopathological biomarkers that may provide prognostic and predictive information for patients with distant metastasis. These may comprise sidedness of the tumor, molecular profile and epigenetic characteristics of the primary tumor and arising metastatic CRC, and early markers reflecting cancer cell resistance in mCRC and biomarkers identified from transcriptome. This review discusses current stage in employment of these biomarkers in clinical practice as well as summarizes current experience in identifying predictive biomarkers in mCRC treatment.
Collapse
Affiliation(s)
- Stanislav Filip
- Department of Oncology and Radiotherapy, Charles University, Faculty of Medicine in Hradec Kralove, Šimkova 870, 50001 Hradec Králové, Czech Republic; (J.P.); (O.K.); (S.J.)
| | - Veronika Vymetalkova
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic; (V.V.); (L.V.); (K.C.)
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, 32300 Pilsen, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 12800 Prague, Czech Republic
| | - Jiri Petera
- Department of Oncology and Radiotherapy, Charles University, Faculty of Medicine in Hradec Kralove, Šimkova 870, 50001 Hradec Králové, Czech Republic; (J.P.); (O.K.); (S.J.)
| | - Ludmila Vodickova
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic; (V.V.); (L.V.); (K.C.)
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, 32300 Pilsen, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 12800 Prague, Czech Republic
| | - Ondrej Kubecek
- Department of Oncology and Radiotherapy, Charles University, Faculty of Medicine in Hradec Kralove, Šimkova 870, 50001 Hradec Králové, Czech Republic; (J.P.); (O.K.); (S.J.)
| | - Stanislav John
- Department of Oncology and Radiotherapy, Charles University, Faculty of Medicine in Hradec Kralove, Šimkova 870, 50001 Hradec Králové, Czech Republic; (J.P.); (O.K.); (S.J.)
| | - Filip Cecka
- Department of Surgery, University Hospital in Hradec Kralove, Sokolská 581, 50005 Hradec Králové, Czech Republic;
| | - Marketa Krupova
- The Fingerland Department of Pathology, University Hospital in Hradec Kralove, Sokolská 581, 50005 Hradec Králové, Czech Republic; (M.K.); (M.M.)
| | - Monika Manethova
- The Fingerland Department of Pathology, University Hospital in Hradec Kralove, Sokolská 581, 50005 Hradec Králové, Czech Republic; (M.K.); (M.M.)
| | - Klara Cervena
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic; (V.V.); (L.V.); (K.C.)
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 12800 Prague, Czech Republic
| | - Pavel Vodicka
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic; (V.V.); (L.V.); (K.C.)
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, 32300 Pilsen, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 12800 Prague, Czech Republic
| |
Collapse
|
8
|
Szafarowski T, Sierdziński J, Ludwig N, Głuszko A, Filipowska A, Szczepański MJ. Assessment of cancer stem cell marker expression in primary head and neck squamous cell carcinoma shows prognostic value for aldehyde dehydrogenase (ALDH1A1). Eur J Pharmacol 2019; 867:172837. [PMID: 31811857 DOI: 10.1016/j.ejphar.2019.172837] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/26/2019] [Accepted: 11/29/2019] [Indexed: 12/21/2022]
Abstract
Cancer stem cells (CSCs) play a key role in carcinogenesis and progression of head and neck squamous cell carcinomas (HNSCC). The most common markers indicating for CSCs are: CD44, CD24, CD133, ALDH1A1. Our objective was to evaluate the prognostic potential of CSC markers in HNSCC. The study included 49 patients treated for primary HNSCC, 11 patients with upper respiratory tract epithelial dysplasia and 12 subjects with the normal pharyngeal mucosa as a control group. The frequency and expression levels of the four CSC markers were assessed by immunohistochemistry. Univariate and multivariate analyses were used to correlate CSC expression levels with tumor stage, lymph node metastases or overall survival (OS). CD44, CD24, CD133, ALDH1A1 were widely expressed in tumors, whereas CD44 was found to be higher in cancer tissue (P = 0.001). ALDH1A1 expression levels were found to be significantly higher in T3-T4 tumors vs. T1-T2 tumors (P = 0.05). Lymph node metastases had significantly higher expression levels of CD24 (P = 0.01) and CD133 (P < 0.05) than primary tumors. Multifactorial analysis revealed that overall survival (OS) for patients with ALDH1A1 negative tumors was 5.25 times higher than for patients with ALDH1A1 positive (ALDH1A1+) tumors (P = 0.01). On univariate and multivariate analysis, only ALDH1A1 positivity had a significant effect on OS of HNSCC patients (HR = 2.47 for P = 0.02). Immunohistochemistry-based assessments of CSC marker expression in HNSCC has significant predictive implications for patients with HNSCC. The frequency of CSCs in the tumor, specifically of ALDH1A1+ cells correlated with five-year OS in these patients.
Collapse
Affiliation(s)
- Tomasz Szafarowski
- Department of Otolaryngology, Faculty of Medicine and Dentistry, Medical University of Warsaw, Stępińska 19/25 Str., 00-739, Warsaw, Poland.
| | - Janusz Sierdziński
- Department of Medical Informatics and Telemedicine, Medical University of Warsaw, Litewska 14/16 Str., 00-581, Warsaw, Poland.
| | - Nils Ludwig
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA; UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA.
| | - Alicja Głuszko
- Department of Biochemistry, First Faculty of Medicine, Medical University of Warsaw, Banacha 1 Str., 02-097, Warsaw, Poland.
| | - Anna Filipowska
- Department of Biosensors and Processing of Biomedical Signals, Silesian University of Technology, Roosevelta 40 Str., 41-800, Zabrze, Poland.
| | - Mirosław J Szczepański
- Department of Biochemistry, First Faculty of Medicine, Medical University of Warsaw, Banacha 1 Str., 02-097, Warsaw, Poland.
| |
Collapse
|
9
|
Liskova A, Kubatka P, Samec M, Zubor P, Mlyncek M, Bielik T, Samuel SM, Zulli A, Kwon TK, Büsselberg D. Dietary Phytochemicals Targeting Cancer Stem Cells. Molecules 2019; 24:molecules24050899. [PMID: 30836718 PMCID: PMC6429493 DOI: 10.3390/molecules24050899] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 02/25/2019] [Accepted: 02/28/2019] [Indexed: 12/11/2022] Open
Abstract
There is an increasing awareness of the importance of a diet rich in fruits and vegetables for human health. Cancer stem cells (CSCs) are characterized as a subpopulation of cancer cells with aberrant regulation of self-renewal, proliferation or apoptosis leading to cancer progression, invasiveness, metastasis formation, and therapy resistance. Anticancer effects of phytochemicals are also directed to target CSCs. Here we provide a comprehensive review of dietary phytochemicals targeting CSCs. Moreover, we evaluate and summarize studies dealing with effects of dietary phytochemicals on CSCs of various malignancies in preclinical and clinical research. Dietary phytochemicals have a significant impact on CSCs which may be applied in cancer prevention and treatment. However, anticancer effects of plant derived compounds have not yet been fully investigated in clinical research.
Collapse
Affiliation(s)
- Alena Liskova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, 03601 Bratislava, Slovakia.
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, 03601 Bratislava, Slovakia.
| | - Marek Samec
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, 03601 Bratislava, Slovakia.
| | - Pavol Zubor
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, 03601 Bratislava, Slovakia.
| | - Milos Mlyncek
- Department of Obstetrics and Gynecology Faculty Hospital Nitra Constantine the Philosopher University, 949 01 Nitra, Slovakia.
| | - Tibor Bielik
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, 03601 Bratislava, Slovakia.
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha 24144, Qatar.
| | - Anthony Zulli
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3011, Australia.
| | - Taeg Kyu Kwon
- Department of Immunology and School of Medicine, Keimyung University, Dalseo-Gu, Daegu 426 01, Korea.
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha 24144, Qatar.
| |
Collapse
|
10
|
Chen W, Allen SG, Qian W, Peng Z, Han S, Li X, Sun Y, Fournier C, Bao L, Lam RH, Merajver SD, Fu J. Biophysical Phenotyping and Modulation of ALDH+ Inflammatory Breast Cancer Stem-Like Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1802891. [PMID: 30632269 PMCID: PMC6486377 DOI: 10.1002/smll.201802891] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/24/2018] [Indexed: 05/23/2023]
Abstract
Cancer stem-like cells (CSCs) have been shown to initiate tumorigenesis and cancer metastasis in many cancer types. Although identification of CSCs through specific marker expression helps define the CSC compartment, it does not directly provide information on how or why this cancer cell subpopulation is more metastatic or tumorigenic. In this study, the functional and biophysical characteristics of aggressive and lethal inflammatory breast cancer (IBC) CSCs at the single-cell level are comprehensively profiled using multiple microengineered tools. Distinct functional (cell migration, growth, adhesion, invasion and self-renewal) and biophysical (cell deformability, adhesion strength and contractility) properties of ALDH+ SUM149 IBC CSCs are found as compared to their ALDH- non-CSC counterpart, providing biophysical insights into why CSCs has an enhanced propensity to metastasize. It is further shown that the cellular biophysical phenotype can predict and determine IBC cells' tumorigenic ability. SUM149 and SUM159 IBC cells selected and modulated through biophysical attributes-adhesion and stiffness-show characteristics of CSCs in vitro and enhance tumorigenicity in in vivo murine models of primary tumor growth. Overall, the multiparametric cellular biophysical phenotyping and modulation of IBC CSCs yields a new understanding of IBC's metastatic properties and how they might develop and be targeted for therapeutic interventions.
Collapse
Affiliation(s)
- Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA, Department of Biomedical Engineering, New York University, Brooklyn, NY 11201, USA,
| | - Steven G. Allen
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109, USA,
| | - Weiyi Qian
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA
| | - Zifeng Peng
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shuo Han
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xiang Li
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yubing Sun
- Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA 01003, USA
| | - Chelsea Fournier
- Undergraduate Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, USA
| | - Liwei Bao
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Raymond H.W. Lam
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Sofia D. Merajver
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109, USA, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA, Department of Biomedical Engineering, Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA,
| |
Collapse
|
11
|
Namba Y, Sogawa C, Okusha Y, Kawai H, Itagaki M, Ono K, Murakami J, Aoyama E, Ohyama K, Asaumi JI, Takigawa M, Okamoto K, Calderwood SK, Kozaki KI, Eguchi T. Depletion of Lipid Efflux Pump ABCG1 Triggers the Intracellular Accumulation of Extracellular Vesicles and Reduces Aggregation and Tumorigenesis of Metastatic Cancer Cells. Front Oncol 2018; 8:376. [PMID: 30364132 PMCID: PMC6191470 DOI: 10.3389/fonc.2018.00376] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 08/22/2018] [Indexed: 12/21/2022] Open
Abstract
The ATP-binding cassette transporter G1 (ABCG1) is a cholesterol lipid efflux pump whose role in tumor growth has been largely unknown. Our transcriptomics revealed that ABCG1 was powerfully expressed in rapidly metastatic, aggregative colon cancer cells, in all the ABC transporter family members. Coincidently, genetic amplification of ABCG1 is found in 10–35% of clinical samples of metastatic cancer cases. Expression of ABCG1 was further elevated in three-dimensional tumoroids (tumor organoids) within stemness-enhancing tumor milieu, whereas depletion of ABCG1 lowered cellular aggregation and tumoroid growth in vitro as well as hypoxia-inducible factor 1α in cancer cells around the central necrotic areas in tumors in vivo. Notably, depletion of ABCG1 triggered the intracellular accumulation of extracellular vesicles (EVs) and regression of tumoroids. Collectively, these data suggest that ABCG1 plays a crucial role in tumorigenesis in metastatic cancer and that depletion of ABCG1 triggers tumor regression with the accumulation of EVs and their derivatives and cargos, implicating a novel ABCG1-targeting therapeutic strategy by which redundant and toxic substances may be accumulated in tumors leading to their regression.
Collapse
Affiliation(s)
- Yuri Namba
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.,Department of Oral and Maxillofacial Radiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Chiharu Sogawa
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yuka Okusha
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hotaka Kawai
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Mami Itagaki
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kisho Ono
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Jun Murakami
- Advanced Research Center for Oral and Craniofacial Sciences, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.,Department of Oral Diagnosis and Dentomaxillofacial Radiology, Okayama University Hospital, Okayama, Japan
| | - Eriko Aoyama
- Advanced Research Center for Oral and Craniofacial Sciences, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kazumi Ohyama
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Jun-Ichi Asaumi
- Department of Oral and Maxillofacial Radiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masaharu Takigawa
- Advanced Research Center for Oral and Craniofacial Sciences, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kuniaki Okamoto
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Stuart K Calderwood
- Division of Molecular and Cellular Biology, Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Ken-Ichi Kozaki
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takanori Eguchi
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.,Advanced Research Center for Oral and Craniofacial Sciences, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
12
|
High NRF2 level mediates cancer stem cell-like properties of aldehyde dehydrogenase (ALDH)-high ovarian cancer cells: inhibitory role of all-trans retinoic acid in ALDH/NRF2 signaling. Cell Death Dis 2018; 9:896. [PMID: 30166520 PMCID: PMC6117306 DOI: 10.1038/s41419-018-0903-4] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/13/2018] [Accepted: 07/19/2018] [Indexed: 12/22/2022]
Abstract
Aldehyde dehydrogenase 1A1 (ALDH1A1) is one of cancer stem cell (CSC) markers, and high ALDH1 expression has been related to drug resistance and facilitated tumor growth. In this study, we investigated the potential involvement of nuclear factor erythroid 2-like 2 (NFE2L2/NRF2) in CSC-like properties of ALDH-high ovarian CSCs. Our experimental system, ALDH1A1-high (ALDH-H) subpopulation, was isolated and stabilized using doxorubicin-resistant ovarian cancer A2780 cells. ALDH-H exerted CSC-like properties such as drug resistance, colony/sphere formation, and enhanced tumor growth along with high levels of CSCs markers compared to ALDH1A1-low (ALDH-L). Levels of NRF2 and subsequent target genes substantially increased in ALDH-H cells, and the increase in ALDH1A1 and p62 was associated with NRF2 upregulation. ALDH1A1-silencing blocked increases in NRF2, drug efflux transporters, and p62, along with CSC markers in ALDH-H cells. The inhibition of p62, which was elevated in ALDH-H, suppressed NRF2 activation. High NRF2 level was confirmed in the ALDH1-high subpopulation from colon cancer HCT116 cells. The functional implication of NRF2 activation in ovarian CSCs was verified by two experimental approaches. First, CSC-like properties such as high CSC markers, chemoresistance, colony/sphere formation, and tumor growth were significantly inhibited by NRF2-silencing in ALDH-H cells. Second, all-trans retinoic acid (ATRA) suppressed ALDH1 expression, inhibiting NRF2 activation, which led to the attenuation of CSC-like properties in ALDH-H cells but not in ALDH-L cells. These results provide insight into the molecular basis of the ALDH1A1-mediated development of CSC-like properties such as stress/treatment resistance, and further suggest the therapeutic potential of ATRA in ALDH-high ovarian CSCs.
Collapse
|
13
|
Yuan X, Wei W, Bao Q, Chen H, Jin P, Jiang W. Metformin inhibits glioma cells stemness and epithelial-mesenchymal transition via regulating YAP activity. Biomed Pharmacother 2018; 102:263-270. [PMID: 29567539 DOI: 10.1016/j.biopha.2018.03.031] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/06/2018] [Accepted: 03/06/2018] [Indexed: 12/22/2022] Open
Abstract
This work aims to study the roles and mechanisms of metformin in glioma cells stemness and epithelial-mesenchymal transition. Here, we found that metformin suppressed glioma cells spheroid formation and size, inhibited the expression of glioma stemness-related marker, CD133. Additionally, Metformin attenuated TGF-β-induced epithelial-mesenchymal transition in glioma cells. Mechanistically, metformin inhibited the nuclear abundance of YAP, a key effector of Hippo pathway, subsequently leading to its cytoplasmic retention, and thus reduced YAP transcriptional modulating activity. Importantly, overexpression of a mutant form of YAP (YAP-5SA) attenuated the inhibition of metformin on glioma cells stemness and epithelial-mesenchymal transition. Thus, metformin inhibits glioma cells stemness and epithelial-mesenchymal transition via regulating YAP activity.
Collapse
Affiliation(s)
- Xuesong Yuan
- Department of Neurosurgery, Changzhou Wujin People's Hospital, The Affliated Hospital of Jiangsu University, No.2 Yongning North Road, Changzhou, 213002, China
| | - Wenfeng Wei
- Department of Neurosurgery, Changzhou Wujin People's Hospital, The Affliated Hospital of Jiangsu University, No.2 Yongning North Road, Changzhou, 213002, China.
| | - Qing Bao
- Department of Neurosurgery, Changzhou Wujin People's Hospital, The Affliated Hospital of Jiangsu University, No.2 Yongning North Road, Changzhou, 213002, China
| | - Hongchun Chen
- Department of Neurosurgery, Changzhou Wujin People's Hospital, The Affliated Hospital of Jiangsu University, No.2 Yongning North Road, Changzhou, 213002, China
| | - Peng Jin
- Department of Neurosurgery, Changzhou Wujin People's Hospital, The Affliated Hospital of Jiangsu University, No.2 Yongning North Road, Changzhou, 213002, China
| | - Wenqing Jiang
- Department of Neurosurgery, Changzhou Wujin People's Hospital, The Affliated Hospital of Jiangsu University, No.2 Yongning North Road, Changzhou, 213002, China
| |
Collapse
|
14
|
ΔNp63 drives metastasis in breast cancer cells via PI3K/CD44v6 axis. Oncotarget 2018; 7:54157-54173. [PMID: 27494839 PMCID: PMC5342332 DOI: 10.18632/oncotarget.11022] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 07/22/2016] [Indexed: 01/03/2023] Open
Abstract
P63 is a transcription factor belonging to the family of p53, essential for the development and differentiation of epithelia. In recent years, it has become clear that altered expression of the different isoforms of this gene can play an important role in carcinogenesis. The p63 gene encodes for two main isoforms known as TA and ΔN p63 with different functions. The role of these different isoforms in sustaining tumor progression and metastatic spreading however has not entirely been clarified. Here we show that breast cancer initiating cells express ΔNp63 isoform that supports a more mesenchymal phenotype associated with a higher tumorigenic and metastatic potential. On the contrary, the majority of cells within the tumor appears to express predominantly TAp63 isoform. While ΔNp63 exerts its effects by regulating a PI3K/CD44v6 pathway, TAp63 modulates this pathway in an opposite fashion. As a result, tumorigenicity and invasive capacity of breast cancer cells is a balance of the two isoforms. Finally, we found that tumor microenvironmental cytokines significantly contribute to the establishment of breast cancer cell phenotype by positively regulating ΔNp63 and CD44v6 expression.
Collapse
|
15
|
Kim YS, Lee HJ, Park JM, Han YM, Kangwan N, Oh JY, Lee DY, Hahm KB. Targeted molecular ablation of cancer stem cells for curing gastrointestinal cancers. Expert Rev Gastroenterol Hepatol 2017; 11:1059-1070. [PMID: 28707966 DOI: 10.1080/17474124.2017.1356224] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Abundance of the ATPase-binding cassette (ABC) transporters and deranged self-renewal pathways characterize the presence of cancer stem cells (CSCs) in gastrointestinal cancers (GI cancers), which play crucial roles in tumorigenesis, chemotherapy resistance, tumor recurrence, and cancer metastasis. Therefore, in order to ensure high cure rates, chemoquiescence, CSCs should be ablated. Recent advances in either understanding CSCs or biomarker identification enable scientists to develop techniques for ablating CSCs and clinicians to provide cancer cure, especially in GI cancers characterized by inflammation-driven carcinogenesis. Areas covered: A novel approach to ablate CSCs in GI cancers, including esophageal, gastric, and colon cancers, is introduced along with explored underlying molecular mechanisms. Expert commentary: Though CSC ablation is still in the empirical stages and not in clinical practice, several strategies for ablating CSCs in GI cancers had been published, proton-pump inhibitors (PPIs) that regulate the membrane-bound ABC transporters, which underlie drug resistance; chloroquine (CQ) that inhibits autophagy, which is responsible for tumor survival; Hedgehog/Wnt/Notch inhibitors that influence the underlying stem-cell growth, and some natural products including Korean red ginseng, cancer-preventive kimchi, Artemisia extract, EGCG from green tea, and walnut extracts.
Collapse
Affiliation(s)
- Yong Seok Kim
- a Department of Biochemistry and Molecular Biology , Hanyang University College of Medicine , Seoul , Korea
| | - Ho Jae Lee
- b Department of Biochemistry , Gachon University College of Medicine , Incheon , Korea
| | - Jong-Min Park
- c CHA Cancer Prevention Research Center , CHA University , Seongnam , Korea
| | - Young-Min Han
- c CHA Cancer Prevention Research Center , CHA University , Seongnam , Korea
| | - Napapan Kangwan
- d Division of Physiology, School of Medical Sciences , University of Phayao , Phayao , Thailand
| | | | | | - Ki Baik Hahm
- a Department of Biochemistry and Molecular Biology , Hanyang University College of Medicine , Seoul , Korea.,c CHA Cancer Prevention Research Center , CHA University , Seongnam , Korea.,f Digestive Disease Center , CHA University Bundang Medical Center , Seongnam , Korea
| |
Collapse
|
16
|
Pleniceanu O, Shukrun R, Omer D, Vax E, Kanter I, Dziedzic K, Pode-Shakked N, Mark-Daniei M, Pri-Chen S, Gnatek Y, Alfandary H, Varda-Bloom N, Bar-Lev DD, Bollag N, Shtainfeld R, Armon L, Urbach A, Kalisky T, Nagler A, Harari-Steinberg O, Arbiser JL, Dekel B. Peroxisome proliferator-activated receptor gamma (PPARγ) is central to the initiation and propagation of human angiomyolipoma, suggesting its potential as a therapeutic target. EMBO Mol Med 2017; 9:508-530. [PMID: 28275008 PMCID: PMC5376758 DOI: 10.15252/emmm.201506111] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Angiomyolipoma (AML), the most common benign renal tumor, can result in severe morbidity from hemorrhage and renal failure. While mTORC1 activation is involved in its growth, mTORC1 inhibitors fail to eradicate AML, highlighting the need for new therapies. Moreover, the identity of the AML cell of origin is obscure. AML research, however, is hampered by the lack of in vivo models. Here, we establish a human AML‐xenograft (Xn) model in mice, recapitulating AML at the histological and molecular levels. Microarray analysis demonstrated tumor growth in vivo to involve robust PPARG‐pathway activation. Similarly, immunostaining revealed strong PPARG expression in human AML specimens. Accordingly, we demonstrate that while PPARG agonism accelerates AML growth, PPARG antagonism is inhibitory, strongly suppressing AML proliferation and tumor‐initiating capacity, via a TGFB‐mediated inhibition of PDGFB and CTGF. Finally, we show striking similarity between AML cell lines and mesenchymal stem cells (MSCs) in terms of antigen and gene expression and differentiation potential. Altogether, we establish the first in vivo human AML model, which provides evidence that AML may originate in a PPARG‐activated renal MSC lineage that is skewed toward adipocytes and smooth muscle and away from osteoblasts, and uncover PPARG as a regulator of AML growth, which could serve as an attractive therapeutic target.
Collapse
Affiliation(s)
- Oren Pleniceanu
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Hematology and Cord Blood Bank, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Racheli Shukrun
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dorit Omer
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Einav Vax
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Itamar Kanter
- Faculty of Engineering, Institute of Nanotechnology, Bar-Ilan University, Ramat Gan, Israel
| | - Klaudyna Dziedzic
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Naomi Pode-Shakked
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal Mark-Daniei
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Sara Pri-Chen
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Yehudit Gnatek
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Hadas Alfandary
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Institute of Nephrology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Nira Varda-Bloom
- Division of Hematology and Cord Blood Bank, Sheba Medical Center, Ramat Gan, Israel
| | - Dekel D Bar-Lev
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Naomi Bollag
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Rachel Shtainfeld
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Leah Armon
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Achia Urbach
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Tomer Kalisky
- Faculty of Engineering, Institute of Nanotechnology, Bar-Ilan University, Ramat Gan, Israel
| | - Arnon Nagler
- Division of Hematology and Cord Blood Bank, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Orit Harari-Steinberg
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Jack L Arbiser
- Department of Dermatology, Emory University School of Medicine, Atlanta, GA, USA.,Winship Cancer Institute, Atlanta Veterans Administration Hospital, Atlanta, GA, USA
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel .,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
17
|
Narayan S, Jaiswal AS, Sharma R, Nawab A, Duckworth LV, Law BK, Zajac-Kaye M, George TJ, Sharma J, Sharma AK, Hromas RA. NSC30049 inhibits Chk1 pathway in 5-FU-resistant CRC bulk and stem cell populations. Oncotarget 2017; 8:57246-57264. [PMID: 28915668 PMCID: PMC5593639 DOI: 10.18632/oncotarget.19778] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/20/2017] [Indexed: 01/20/2023] Open
Abstract
The 5-fluorouracil (5-FU) treatment induces DNA damage and stalling of DNA replication forks. These stalled replication forks then collapse to form one sided double-strand breaks, leading to apoptosis. However, colorectal cancer (CRC) stem cells rapidly repair the stalled/collapsed replication forks and overcome treatment effects. Recent evidence suggests a critical role of checkpoint kinase 1 (Chk1) in preventing the replicative stress. Therefore, Chk1 kinase has been a target for developing mono or combination therapeutic agents. In the present study, we have identified a novel orphan molecule NSC30049 (NSC49L) that is effective alone, and in combination potentiates 5-FU-mediated growth inhibition of CRC heterogeneous bulk and FOLFOX-resistant cell lines in culture with minimal effect on normal colonic epithelial cells. It also inhibits the sphere forming activity of CRC stem cells, and decreases the expression levels of mRNAs of CRC stem cell marker genes. Results showed that NSC49L induces 5-FU-mediated S-phase cell cycle arrest due to increased load of DNA damage and increased γ-H2AX staining as a mechanism of cytotoxicity. The pharmacokinetic analysis showed a higher bioavailability of this compound, however, with a short plasma half-life. The drug is highly tolerated by animals with no pathological aberrations. Furthermore, NSC49L showed very potent activity in a HDTX model of CRC stem cell tumors either alone or in combination with 5-FU. Thus, NSC49L as a single agent or combined with 5-FU can be developed as a therapeutic agent by targeting the Chk1 pathway in 5-FU-resistant CRC heterogeneous bulk and CRC stem cell populations.
Collapse
Affiliation(s)
- Satya Narayan
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA
| | - Aruna S. Jaiswal
- Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Ritika Sharma
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA
| | - Akbar Nawab
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA
| | - Lizette Vila Duckworth
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Brian K. Law
- Department of Pharmacology and Experimental Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | - Maria Zajac-Kaye
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA
| | - Thomas J. George
- Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jay Sharma
- Celprogen, Inc., Torrance, CA 90503, USA
| | - Arun K. Sharma
- Department of Pharmacology, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Robert A. Hromas
- Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
18
|
Yu L, Zhu B, Wu S, Zhou L, Song W, Gong X, Wang D. Evaluation of the correlation of vasculogenic mimicry, ALDH1, KiSS-1, and MACC1 in the prediction of metastasis and prognosis in ovarian carcinoma. Diagn Pathol 2017; 12:23. [PMID: 28253891 PMCID: PMC5335811 DOI: 10.1186/s13000-017-0612-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 02/20/2017] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Recurrence and metastasis are the usual manifestations of treatment failure of epithelial ovarian carcinoma (EOC). Vasculogenic mimicry (VM; blood supply development often seen in highly aggressive cancers), aldehyde dehydrogenase 1 (ALDH1, cancer stem cell biomarker), KiSS-1 (suppressor of tumor metastasis), and metastasis associated in colon cancer-1 (MACC1) are all useful predictive factors for metastasis and prognosis in various cancers. In this study, we analyzed associations among VM, ALDH1, KiSS-1, and MACC1 in EOC, and their respective correlations with clinicopathological characteristics and survival in EOC. METHODS Positive rates of VM, ALDH1, KiSS-1, and MACC1 in 207 whole EOC tissue samples were detected by immunohistochemistry. Patients' clinical data were also collected. RESULTS Levels of VM, ALDH1, and MACC1 were significantly higher, and levels of KiSS-1 significantly lower, in EOC tissues than in benign ovary tumors. Levels of VM, ALDH1, KiSS-1, and MACC1 were associated significantly with tumor/lymph node/metastasis (LNM) grade, implantation, and International Federation of Gynecology and Obstetrics (FIGO) stage, and with patients' overall survival (OS); whereas the KiSS-1+ subgroup had significantly longer OS than did the KiSS-1- subgroup. In multivariate analysis, high VM, ALDH1 or MACC1 levels, FIGO stage, implantation and low KiSS-1 levels were independently associated with shorter OS in patients with EOC. CONCLUSIONS VM and expressions of ALDH1, KiSS-1, and MACC1 represent promising markers for metastasis and prognosis, and potential therapeutic targets for EOC.
Collapse
Affiliation(s)
- Lan Yu
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China
| | - Bo Zhu
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China
| | - Shiwu Wu
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China.
| | - Lei Zhou
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China
| | - Wenqing Song
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China
| | - Xiaomeng Gong
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China
| | - Danna Wang
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China
| |
Collapse
|
19
|
Li Z, Yin S, Zhang L, Liu W, Chen B, Xing H. Clinicopathological characteristics and prognostic value of cancer stem cell marker CD133 in breast cancer: a meta-analysis. Onco Targets Ther 2017; 10:859-870. [PMID: 28243121 PMCID: PMC5317305 DOI: 10.2147/ott.s124733] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background The association of CD133 overexpression with clinicopathological significance and prognosis in patients with breast cancer remains controversial. We thus performed a meta-analysis to evaluate the role of CD133 expression in the development and prognosis of breast cancer. Methods The databases PubMed, Embase, and Cochrane Library (updated to August 1, 2016) were searched. Pooled odds ratios (ORs) or hazard ratios (HRs) with 95% confidence intervals (95% CI) were used to evaluate the impact of CD133 expression on clinicopathological features, overall survival, and disease-free survival. Results A total of 1,734 patients from 13 studies were subject to final analysis. The results showed a significant association between overexpression of CD133 and estrogen receptor status (OR 0.35, 95% CI 0.18–0.70), progesterone receptor status (OR 0.56, 95% CI 0.43–0.74), human epidermal growth factor-2 status (OR 1.81, 95% CI 1.33–2.45), lymph node metastasis (OR 1.98, 95% CI 1.34–2.92), and tumor histological grade (OR 1.79, 95% CI 1.26–2.54) in breast cancer. However, no significant correlation was found between upregulation of CD133 expression and onset age (OR 1.03, 95% CI 0.70–1.53) or tumor size (OR 1.29, 95% CI 0.80–2.09). Moreover, CD133-positive breast cancer patients had a higher risk of mortality (HR 1.91, 95% CI 1.21–3.03) and disease progression (HR 2.70, 95% CI 1.05–6.95). Conclusion This meta-analysis suggested that CD133 might be a predictor of clinical outcomes as well as prognosis and could be a potentially new gene therapy target for breast cancer patients.
Collapse
Affiliation(s)
- Zhan Li
- Department of Breast Surgery
| | - Songcheng Yin
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning
| | | | | | - Bo Chen
- Department of Breast Surgery
| | - Hua Xing
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| |
Collapse
|
20
|
Hedgehog Pathway Inhibition Hampers Sphere and Holoclone Formation in Rhabdomyosarcoma. Stem Cells Int 2017; 2017:7507380. [PMID: 28243259 PMCID: PMC5294584 DOI: 10.1155/2017/7507380] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 12/02/2016] [Accepted: 12/28/2016] [Indexed: 01/06/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is the most common type of soft tissue sarcoma in children and can be divided into two main subtypes: embryonal (eRMS) and alveolar (aRMS). Among the cellular heterogeneity of tumors, the existence of a small fraction of cells called cancer stem cells (CSC), thought to be responsible for the onset and propagation of cancer, has been demonstrated in some neoplasia. Although the existence of CSC has been reported for eRMS, their existence in aRMS, the most malignant subtype, has not been demonstrated to date. Given the lack of suitable markers to identify this subpopulation in aRMS, we used cancer stem cell-enriched supracellular structures (spheres and holoclones) to study this subpopulation. This strategy allowed us to demonstrate the capacity of both aRMS and eRMS cells to form these structures and retain self-renewal capacity. Furthermore, cells contained in spheres and holoclones showed significant Hedgehog pathway induction, the inhibition of which (pharmacologic or genetic) impairs the formation of both holoclones and spheres. Our findings point to a crucial role of this pathway in the maintenance of these structures and suggest that Hedgehog pathway targeting in CSC may have great potential in preventing local relapses and metastases.
Collapse
|
21
|
Shaikh MV, Kala M, Nivsarkar M. CD90 a potential cancer stem cell marker and a therapeutic target. Cancer Biomark 2016; 16:301-7. [PMID: 27062695 DOI: 10.3233/cbm-160590] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cancer Stem Cells (CSCs) have been recently identified and their role in carcinogenesis has been ascertained. CSCs have been correlated with high relapse in certain cancers, multiple drug resistance against chemotherapy and metastasis. Several markers such as CD133, CD24, CD44, EpCAM, and CD26 have been identified to isolate and characterize CSCs. None of these markers or their combinations are universal in nature and can be used to isolate CSCs from all types of cancer. CD90 is one such marker whose expression has been extensively studied in recent years. CD90+ cells have been isolated from several types of tumors and shown to exhibit cardinal properties of CSCs such as proliferation, differentiation, spheroid formation, metastasis and ability to form tumor xenograft in immunodeficient mice. It is also found to be co-expressed with several other CSC markers. CD90 is therefore, suggested as a candidate marker as well as a potential therapeutic target for elimination of CSCs.
Collapse
Affiliation(s)
- Muhammad Vaseem Shaikh
- Department of Pharmacology and Toxicology, B. V. Patel Pharmaceutical Education and Research Development Centre, S. G. Highway, Thaltej, Ahmedabad, Gujarat, India.,Faculty of Pharmacy, NIRMA University, Sarkhej-Gandhinagar Highway, Gota, Ahmedabad, Gujarat, India
| | - Manika Kala
- Department of Pharmacology and Toxicology, B. V. Patel Pharmaceutical Education and Research Development Centre, S. G. Highway, Thaltej, Ahmedabad, Gujarat, India.,Faculty of Pharmacy, NIRMA University, Sarkhej-Gandhinagar Highway, Gota, Ahmedabad, Gujarat, India
| | - Manish Nivsarkar
- Department of Pharmacology and Toxicology, B. V. Patel Pharmaceutical Education and Research Development Centre, S. G. Highway, Thaltej, Ahmedabad, Gujarat, India
| |
Collapse
|
22
|
Charni M, Aloni-Grinstein R, Molchadsky A, Rotter V. p53 on the crossroad between regeneration and cancer. Cell Death Differ 2016; 24:8-14. [PMID: 27768121 PMCID: PMC5260496 DOI: 10.1038/cdd.2016.117] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 09/18/2016] [Accepted: 09/19/2016] [Indexed: 12/19/2022] Open
Abstract
Regeneration and tumorigenesis share common molecular pathways, nevertheless the outcome of regeneration is life, whereas tumorigenesis leads to death. Although the process of regeneration is strictly controlled, malignant transformation is unrestrained. In this review, we discuss the involvement of TP53, the major tumor-suppressor gene, in the regeneration process. We point to the role of p53 as coordinator assuring that regeneration will not shift to carcinogenesis. The fluctuation in p53 activity during the regeneration process permits a tight control. On one hand, its inhibition at the initial stages allows massive proliferation, on the other its induction at advanced steps of regeneration is essential for preservation of robustness and fidelity of the regeneration process. A better understanding of the role of p53 in regulation of regeneration may open new opportunities for implementation of TP53-based therapies, currently available for cancer patients, in regenerative medicine.
Collapse
Affiliation(s)
- Meital Charni
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ronit Aloni-Grinstein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Alina Molchadsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Varda Rotter
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
23
|
All-trans retinoic acid targets gastric cancer stem cells and inhibits patient-derived gastric carcinoma tumor growth. Oncogene 2016; 35:5619-5628. [PMID: 27157616 DOI: 10.1038/onc.2016.87] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/02/2016] [Accepted: 02/12/2016] [Indexed: 02/07/2023]
Abstract
Gastric carcinoma is the third leading cause of cancer-related death worldwide. This cancer, most of the time metastatic, is essentially treated by surgery associated with conventional chemotherapy, and has a poor prognosis. The existence of cancer stem cells (CSC) expressing CD44 and a high aldehyde dehydrogenase (ALDH) activity has recently been demonstrated in gastric carcinoma and has opened new perspectives to develop targeted therapy. In this study, we evaluated the effects of all-trans-retinoic acid (ATRA) on CSCs in human gastric carcinoma. ATRA effects were evaluated on the proliferation and tumorigenic properties of gastric carcinoma cells from patient-derived tumors and cell lines in conventional 2D cultures, in 3D culture systems (tumorsphere assay) and in mouse xenograft models. ATRA inhibited both tumorspheres initiation and growth in vitro, which was associated with a cell-cycle arrest through the upregulation of cyclin-dependent kinase (CDK) inhibitors and the downregulation of cell-cycle progression activators. More importantly, ATRA downregulated the expression of the CSC markers CD44 and ALDH as well as stemness genes such as Klf4 and Sox2 and induced differentiation of tumorspheres. Finally, 2 weeks of daily ATRA treatment were sufficient to inhibit gastric tumor progression in vivo, which was associated with a decrease in CD44, ALDH1, Ki67 and PCNA expression in the remaining tumor cells. Administration of ATRA appears to be a potent strategy to efficiently inhibit tumor growth and more importantly to target gastric CSCs in both intestinal and diffuse types of gastric carcinoma.
Collapse
|
24
|
Ullah N, Liaqat S, Fatima S, Zehra F, Anwer M, Sadiq M. Stem cells and cancer: A review. ASIAN PACIFIC JOURNAL OF TROPICAL DISEASE 2016. [DOI: 10.1016/s2222-1808(15)61057-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
25
|
Hu Y, Li S. Survival regulation of leukemia stem cells. Cell Mol Life Sci 2016; 73:1039-50. [PMID: 26686687 PMCID: PMC11108378 DOI: 10.1007/s00018-015-2108-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 11/30/2015] [Accepted: 12/01/2015] [Indexed: 02/05/2023]
Abstract
Leukemia stem cells (LSCs) are a subpopulation cells at the apex of hierarchies in leukemia cells and responsible for disease continuous propagation. In this article, we discuss some cellular and molecular components, which are critical for LSC survival. These components include intrinsic signaling pathways and extrinsic microenvironments. The intrinsic signaling pathways to be discussed include Wnt/β-catenin signaling, Hox genes, Hh pathway, Alox5, and some miRNAs, which have been shown to play important roles in regulating LSC survival and proliferation. The extrinsic components to be discussed include selectins, CXCL12/CXCR4, and CD44, which involve in LSC homing, survival, and proliferation by affecting bone marrow microenvironment. Potential strategies for eradicating LSCs will also discuss.
Collapse
Affiliation(s)
- Yiguo Hu
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, No. 17, The Third Part Renmin South Road, Chengdu, 610041, Sichuan, China.
| | - Shaoguang Li
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
26
|
Cancer Stem Cells and Macrophages: Implications in Tumor Biology and Therapeutic Strategies. Mediators Inflamm 2016; 2016:9012369. [PMID: 26980947 PMCID: PMC4769767 DOI: 10.1155/2016/9012369] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 12/31/2015] [Indexed: 12/28/2022] Open
Abstract
Cancer stem cells (CSCs) are a unique subset of cells within tumors with stemlike properties that have been proposed to be key drivers of tumor initiation and progression. CSCs are functionally defined by their unlimited self-renewal capacity and their ability to initiate tumor formation in vivo. Like normal stem cells, CSCs exist in a cellular niche comprised of numerous cell types including tumor-associated macrophages (TAMs) which provides a unique microenvironment to protect and promote CSC functions. TAMs provide pivotal signals to promote CSC survival, self-renewal, maintenance, and migratory ability, and in turn, CSCs deliver tumor-promoting cues to TAMs that further enhance tumorigenesis. Studies in the last decade have aimed to understand the molecular mediators of CSCs and TAMs, and recent advances have begun to elucidate the complex cross talk that occurs between these two cell types. In this review, we discuss the molecular interactions that define CSC-TAM cross talk at each stage of tumor progression and examine the clinical implications of targeting these interactions.
Collapse
|
27
|
Tissue Regeneration in the Chronically Inflamed Tumor Environment: Implications for Cell Fusion Driven Tumor Progression and Therapy Resistant Tumor Hybrid Cells. Int J Mol Sci 2015; 16:30362-81. [PMID: 26703575 PMCID: PMC4691180 DOI: 10.3390/ijms161226240] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/07/2015] [Accepted: 12/09/2015] [Indexed: 12/19/2022] Open
Abstract
The biological phenomenon of cell fusion in a cancer context is still a matter of controversial debates. Even though a plethora of in vitro and in vivo data have been published in the past decades the ultimate proof that tumor hybrid cells could originate in (human) cancers and could contribute to the progression of the disease is still missing, suggesting that the cell fusion hypothesis is rather fiction than fact. However, is the lack of this ultimate proof a valid argument against this hypothesis, particularly if one has to consider that appropriate markers do not (yet) exist, thus making it virtually impossible to identify a human tumor cell clearly as a tumor hybrid cell. In the present review, we will summarize the evidence supporting the cell fusion in cancer concept. Moreover, we will refine the cell fusion hypothesis by providing evidence that cell fusion is a potent inducer of aneuploidy, genomic instability and, most likely, even chromothripsis, suggesting that cell fusion, like mutations and aneuploidy, might be an inducer of a mutator phenotype. Finally, we will show that "accidental" tissue repair processes during cancer therapy could lead to the origin of therapy resistant cancer hybrid stem cells.
Collapse
|
28
|
Abstract
Over the last decade, compelling evidence has shown that cancer stem cells (CSCs) exist in a variety of malignancies. The conventional method for anticancer therapy involves targeting only the proliferating mitotic cells, sparing the slow-cycling cells that eventually evade chemotherapy and become a source of post therapy relapses. With the increasing awareness of CSCs supported by sophisticated experimental evidence, therapeutic strategies today are aimed at selectively identifying and targeting CSCs using biomarkers. The ability to identify CSCs allows targeted elimination of these cancer-initiating cells. Herein, we discuss CSC markers in the context of different types of cancers, their significance in selectively identifying CSCs and the therapeutic implications of using these biomarkers to prevent invasion and metastasis of cancer.
Collapse
Affiliation(s)
- Madhura Murar
- Symbiosis School of Biomedical Sciences (SSBS), Symbiosis International University (SIU), Symbiosis Knowledge Village, Gram: Lavale, Taluka: Mulshi, District: Pune 412115, Maharashtra, India
| | - Anuradha Vaidya
- Symbiosis School of Biomedical Sciences (SSBS), Symbiosis International University (SIU), Symbiosis Knowledge Village, Gram: Lavale, Taluka: Mulshi, District: Pune 412115, Maharashtra, India
| |
Collapse
|
29
|
Walther V, Alison MR. Cell lineage tracing in human epithelial tissues using mitochondrial DNA mutations as clonal markers. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 5:103-17. [PMID: 26302049 DOI: 10.1002/wdev.203] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 06/20/2015] [Accepted: 07/01/2015] [Indexed: 12/29/2022]
Abstract
The study of cell lineages through heritable genetic lineage tracing is well established in experimental animals, particularly mice. While such techniques are not feasible in humans, we have taken advantage of the fact that the mitochondrial genome is highly prone to nonpathogenic mutations and such mutations can be used as clonal markers to identify stem cell derived clonal populations in human tissue sections. A mitochondrial DNA (mtDNA) mutation can spread by a stochastic process through the several copies of the circular genome in a single mitochondrion, and then through the many mitochondria in a single cell, a process called 'genetic drift.' This process takes many years and so is likely to occur only in stem cells, but once established, the fate of stem cell progeny can be followed. A cell having at least 80% of its mtDNA genomes bearing the mutation results in a demonstrable deficiency in mtDNA-encoded cytochrome c oxidase (CCO), optimally detected in frozen tissue sections by dual-color histochemistry, whereby CCO activity stains brown and CCO deficiency is highlighted by subsequent succinate dehydrogenase activity, staining the CCO-deficient areas blue. Cells with CCO deficiency can be laser captured and subsequent mtDNA sequencing can ascertain the nature of the mutation. If all cells in a CCO-deficient area have an identical mutation, then a clonal population has been identified; the chances of the same mutation initially arising in separate cells are highly improbable. The technique lends itself to the study of both normal epithelia and can answer several questions in tumor biology. WIREs Dev Biol 2016, 5:103-117. doi: 10.1002/wdev.203 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Viola Walther
- Centre for Tumour Biology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Malcolm R Alison
- Centre for Tumour Biology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
30
|
miRNA therapy targeting cancer stem cells: a new paradigm for cancer treatment and prevention of tumor recurrence. Ther Deliv 2015; 6:323-37. [PMID: 25853308 DOI: 10.4155/tde.14.122] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) are a small subpopulation of cells within tumors that retain the properties of self-renewal and tumorigenicity in vivo. Although CSCs have been reported in multiple cancers, the regulation of CSCs has not been described at the molecular level. miRNAs are endogenous small noncoding RNAs that post-transcriptionally regulate the expression of their target genes via RNA interference and are involved in almost all cellular processes. Since aberrant miRNA expression occurs in CSCs, such dysregulated miRNAs may be promising therapeutic targets. In this review, we summarize the current knowledge regarding miRNAs that regulate CSC properties and discuss an in vivo delivery system for synthetic miRNA mimics and miRNA inhibitors for the development of innovative miRNA therapy against CSCs.
Collapse
|
31
|
Lee E, Yang J, Ku M, Kim NH, Park Y, Park CB, Suh JS, Park ES, Yook JI, Mills GB, Huh YM, Cheong JH. Metabolic stress induces a Wnt-dependent cancer stem cell-like state transition. Cell Death Dis 2015; 6:e1805. [PMID: 26136078 PMCID: PMC4650724 DOI: 10.1038/cddis.2015.171] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Revised: 04/17/2015] [Accepted: 04/21/2015] [Indexed: 12/21/2022]
Abstract
Reciprocal interactions between cancer cells and the tumor microenvironment drive multiple clinically significant behaviors including dormancy, invasion, and metastasis as well as therapy resistance. These microenvironment-dependent phenotypes share typical characteristics with cancer stem cells (CSC). However, it is poorly understood how metabolic stress in the confined tumor microenvironment contributes to the emergence and maintenance of CSC-like phenotypes. Here, we demonstrate that chronic metabolic stress (CMS) in a long-term nutrient deprivation induces a Wnt-dependent phenoconversion of non-stem cancer cells toward stem-like state and this is reflected in the transcriptome analysis. Addition of Wnt3a as well as transfection of dominant-negative Tcf4 establishes an obligatory role for the Wnt pathway in the acquisition of CSC-like characteristics in response to metabolic stress. Furthermore, systematic characterization for multiple single cell-derived clones and negative enrichment of CD44+/ESA+ stem-like cancer cells, all of which recapitulate stem-like cancer characteristics, suggest stochastic adaptation rather than selection of pre-existing subclones. Finally, CMS in the tumor microenvironment can drive a CSC-like phenoconversion of non-stem cancer cells through stochastic state transition dependent on the Wnt pathway. These findings contribute to an understanding of the metabolic stress-driven dynamic transition of non-stem cancer cells to a stem-like state in the tumor metabolic microenvironment.
Collapse
Affiliation(s)
- E Lee
- Department of Radiology, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
- Nanomedical National Core Research Center, Yonsei University, Seoul 120-749, Republic of Korea
| | - J Yang
- Department of Radiology, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
- Yonsei-KRIBB Medical Convergence Research Institute, Yonsei University Health System, Seoul, Korea
| | - M Ku
- Department of Radiology, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul 120-752, Republic of Korea
| | - N H Kim
- Department of Oral pathology, Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul 120-752, Republic of Korea
| | - Y Park
- Department of Radiology, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - C B Park
- Department of Oral pathology, Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul 120-752, Republic of Korea
| | - J-S Suh
- Department of Radiology, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
- Yonsei-KRIBB Medical Convergence Research Institute, Yonsei University Health System, Seoul, Korea
- Severance Biomedical Science Institute (SBSI), Seoul 120-752, Republic of Korea
| | - E S Park
- Yonsei-KRIBB Medical Convergence Research Institute, Yonsei University Health System, Seoul, Korea
| | - J I Yook
- Department of Oral pathology, Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul 120-752, Republic of Korea
| | - G B Mills
- Department of Systems Biology, MD Anderson Cancer Center, Houston, TX, USA
| | - Y-M Huh
- Department of Radiology, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
- Yonsei-KRIBB Medical Convergence Research Institute, Yonsei University Health System, Seoul, Korea
- Severance Biomedical Science Institute (SBSI), Seoul 120-752, Republic of Korea
| | - J-H Cheong
- Severance Biomedical Science Institute (SBSI), Seoul 120-752, Republic of Korea
- Department of Surgery, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
- Department of Biochemistry & Molecular Biology, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| |
Collapse
|
32
|
Kapucuoğlu N, Bozkurt KK, Başpınar Ş, Koçer M, Eroğlu HE, Akdeniz R, Akçil M. The clinicopathological and prognostic significance of CD24, CD44, CD133, ALDH1 expressions in invasive ductal carcinoma of the breast: CD44/CD24 expression in breast cancer. Pathol Res Pract 2015; 211:740-7. [PMID: 26298632 DOI: 10.1016/j.prp.2015.05.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 03/08/2015] [Accepted: 05/27/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND Recently, there are several studies about cancer stem cells (CSC), indicating that they are the cells that initiate the tumor, provide progression, metastasis and responsible for the aggressive tumor behavior. MATERIALS AND METHODS The purpose of this study is to investigate the expressions of CD24, CD44, their different combinations, ALDH1 and CD133 in invasive ductal carcinoma. Their relationships with clinicopathologic parameters, such as tumor grade, lymphovascular invasion, tumor size, axillary lymph node involvement, stage, hormone receptors, HER2 expression, basal like tumors, triple negative status and prognosis were also investigated. Tissue microarray method was used to investigate ımmunohistochemical CD24, CD44, ALDH1 and CD133 expressions in 105 invasive ductal carcinoma cases. RESULTS CD133 expression was significantly associated with tumor size (p=0.023) and stage (p=0.009). CD133 expression was decreased in tumors with larger tumor size, higher stage and lymphovascular invasion. CD133 expression was positively correlated with CD44 (r=0.212, p=0.032) and CD44(+)/CD24(+) (r=0.202, p=0.040) expressions. CD44, CD24 and ALDH1 expressions showed no significant relationship and correlation with clinicopathologic features. There was a significant relationship (p=0.048) between CD44(+)/CD24(-/low) phenotype and basal like tumors. EGFR expression was positively correlated with CD44(+)/CD24(-/low) phenotype (r=0.211, p=0.036). CONCLUSIONS Basal like tumors are enriched for CSCs with CD44(+)/CD24(-/low) phenotype. CD133 can detect a different population of CSC in breast carcinoma.
Collapse
Affiliation(s)
- Nilgün Kapucuoğlu
- Süleyman Demirel University, Faculty of Medicine, Department of Pathology, 32260 Isparta, Turkey.
| | - Kemal Kürşat Bozkurt
- Süleyman Demirel University, Faculty of Medicine, Department of Pathology, 32260 Isparta, Turkey.
| | - Şirin Başpınar
- Süleyman Demirel University, Faculty of Medicine, Department of Pathology, 32260 Isparta, Turkey.
| | - Murat Koçer
- Süleyman Demirel University, Faculty of Medicine, Department of Medical Oncology, 32260 Isparta, Turkey.
| | - Hasan Erol Eroğlu
- Süleyman Demirel University, Faculty of Medicine, Department of General Surgery, 32260 Isparta, Turkey.
| | - Raşit Akdeniz
- Süleyman Demirel University, Faculty of Medicine, Department of Pathology, 32260 Isparta, Turkey.
| | - Mehtap Akçil
- Başkent University, Faculty of Arts, Department of Biostatistics, Ankara, Turkey.
| |
Collapse
|
33
|
Zhou H, Hu YU, Wang W, Mao Y, Zhu J, Zhou B, Sun J, Zhang X. Expression of Oct-4 is significantly associated with the development and prognosis of colorectal cancer. Oncol Lett 2015; 10:691-696. [PMID: 26622555 DOI: 10.3892/ol.2015.3269] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 05/07/2015] [Indexed: 01/03/2023] Open
Abstract
Octamer-binding transcription factor 4 (Oct-4), is an essential transcription factor, which is required for pluripotency and self-renewal in embryonic stem cells and germ cells. It is also involved in maintaining cancer stem-like properties in certain types of tumor, and is an important biomarker for cancer stem cells. The present study investigated whether Oct-4 expression was associated with colorectal cancer (CRC). In order to achieve this, primary CRC tissues, matched non-tumor tissues and benign polyp tissues, representing different stages of carcinogenesis, were obtained, and Oct-4 expression was analyzed using reverse transcription-quantitative polymerase chain reaction, flow cytometry analysis and immunohistochemistry. Furthermore, the medical records of patients with CRC were reviewed, and clinicopathological analysis was performed in order to assess the association between Oct-4 expression and certain clinicopathological parameters. It was shown that the transcription and translation of Oct-4 increased in a stepwise manner, from non-tumor to benign polyp tissues, and from benign polyps to CRC tissues. Oct-4 expression in CRC was significantly correlated with histological grade (P=0.007), lymph node metastasis (P=0.027), distant metastasis (P=0.017) and TNM stage (P=0.041). Kaplan-Meier survival curve analysis demonstrated that Oct-4+ cases had a shorter median survival time (37.0 months) compared with Oct-4- cases (76.0 months; P=0.001). These results indicated that aberrant expression of Oct-4 may be involved in the development of CRC. Thus, Oct-4 may be a biomarker for the prediction, diagnosis or assessment of prognosis in CRC, in addition to a potential target for the treatment of this disease.
Collapse
Affiliation(s)
- Huan Zhou
- Medical Biotechnology Institute of Soochow University, Soochow University, Suzhou, Jiangsu 215007, P.R. China ; Department of Clinical Laboratory, No. 4 People's Hospital of Wuxi, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| | - Y U Hu
- Department of Clinical Laboratory, No. 4 People's Hospital of Wuxi, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| | - Weipeng Wang
- School of Pharmacy, Medical College, Soochow University, Soochow, Jiangsu 215123, P.R. China
| | - Yong Mao
- Department of Clinical Laboratory, No. 4 People's Hospital of Wuxi, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| | - Jingjie Zhu
- Department of Respiratory Medicine, First Affiliated Hospital of Soochow University, Soochow, Jiangsu 215006, P.R. China
| | - Bin Zhou
- Medical Biotechnology Institute of Soochow University, Soochow University, Suzhou, Jiangsu 215007, P.R. China ; Jiangsu Institute of Clinical Immunology, Suzhou, Jiangsu 215007, P.R. China
| | - Jing Sun
- Medical Biotechnology Institute of Soochow University, Soochow University, Suzhou, Jiangsu 215007, P.R. China ; Jiangsu Stem Cell Key Laboratory, Suzhou, Jiangsu 215007, P.R. China
| | - Xueguang Zhang
- Medical Biotechnology Institute of Soochow University, Soochow University, Suzhou, Jiangsu 215007, P.R. China ; Jiangsu Institute of Clinical Immunology, Suzhou, Jiangsu 215007, P.R. China ; Jiangsu Stem Cell Key Laboratory, Suzhou, Jiangsu 215007, P.R. China
| |
Collapse
|
34
|
Liang X, Graham KA, Johannessen AC, Costea DE, Labeed FH. Human oral cancer cells with increasing tumorigenic abilities exhibit higher effective membrane capacitance. Integr Biol (Camb) 2014; 6:545-54. [PMID: 24663430 DOI: 10.1039/c3ib40255j] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Although cells with tumorigenic/stem cell-like properties have been identified in many cancers, including oral squamous cell carcinoma (OSCC), their isolation and characterisation is still at early stages. The aim of this study is to characterise the electrophysiological properties of OSCC cells with different tumorigenic properties in order to establish if a correlation exists between tumorigenicity and cellular electrical characteristics. MATERIALS AND METHODS Rapid adherence to collagen IV was used as a non-invasive, functional method to isolate subsets of cells with different tumorigenic abilities from one oral dysplastic and three OSCC-derived cell lines. The cell subsets identified and isolated using this method were further investigated using dielectrophoresis, a label-free method to determine their electrophysiological parameters. Cell membrane morphology was investigated using scanning electron microscopy (SEM) and modulated by use of 4-methylumbelliferone (4-MU). RESULTS Rapid adherent cells (RAC) to collagen IV, enriched for increased tumorigenic ability, had significantly higher effective membrane capacitance than middle (MAC) and late (LAC) adherent cells. SEM showed that, in contrast to MAC and LAC, RAC displayed a rough surface, extremely rich in cellular protrusions. Treatment with 4-MU dramatically altered RAC membrane morphology by causing loss of filopodia, and significantly decreased their membrane capacitance, indicating that the highest membrane capacitance found in RAC was due to their cell membrane morphology. CONCLUSION This is the first study showing that OSCC cells with higher tumour formation ability exhibit higher effective membrane capacitance than cells that are less tumorigenic. OSSC cells with different tumorigenic ability possessed different electrophysiological properties mostly due to their differences in the cell membrane morphology. These results suggest that dielectrophoresis could potentially used in the future for reliable, label-free isolation of putative tumorigenic cells.
Collapse
Affiliation(s)
- X Liang
- The Gade Laboratory for Pathology, Department of Clinical Medicine, Faculty of Medicine and Dentistry, University of Bergen, 5021, Bergen, Norway
| | | | | | | | | |
Collapse
|
35
|
Abstract
Recently, the interconversion between differentiated and stem-like cancer cells has been observed. Here, we model the in vitro growth of heterogeneous cell cultures in the presence of interconversion from differentiated cancer cells to cancer stem cells (CSCs), showing that, by targeting only CSC with cytotoxic agents, it is not always possible to eradicate cancer. We have determined the kinetic conditions under which cytotoxic agents in in vitro heterogeneous cultures of cancer cells eradicate cancer. In particular, we have shown that the chemotherapeutic elimination of in vitro cultures of heterogeneous cancer cells is effective only if it targets all cancer cell types, and if the induced death rates for the different subpopulations of cancer cell types are large enough. The quantitative results of the model are compared and validated with experimental data.
Collapse
Affiliation(s)
- Rui Dilão
- University of Lisbon, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisbon, Portugal. Institut des Hautes Études Scientifiques, 35, route de Chartres, F-91440 Bures-sur-Yvette, France
| |
Collapse
|
36
|
Zhao A, Tohidkia MR, Siegel DL, Coukos G, Omidi Y. Phage antibody display libraries: a powerful antibody discovery platform for immunotherapy. Crit Rev Biotechnol 2014; 36:276-89. [PMID: 25394539 DOI: 10.3109/07388551.2014.958978] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Phage display technology (PDT), a combinatorial screening approach, provides a molecular diversity tool for creating libraries of peptides/proteins and discovery of new recombinant therapeutics. Expression of proteins such as monoclonal antibodies (mAbs) on the surface of filamentous phage can permit the selection of high affinity and specificity therapeutic mAbs against virtually any target antigen. Using a number of diverse selection platforms (e.g. solid phase, solution phase, whole cell and in vivo biopannings), phage antibody libraries (PALs) from the start point provides great potential for the isolation of functional mAb fragments with diagnostic and/or therapeutic purposes. Given the pivotal role of PDT in the discovery of novel therapeutic/diagnostic mAbs, in the current review, we provide an overview on PALs and discuss their impact in the advancement of engineered mAbs.
Collapse
Affiliation(s)
- Aizhi Zhao
- a Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania , Philadelphia , PA , USA
| | - Mohammad R Tohidkia
- b Research Center for Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Donald L Siegel
- c Division of Transfusion Medicine, Department of Pathology & Laboratory Medicine , University of Pennsylvania School of Medicine , Philadelphia , PA , USA , and
| | - George Coukos
- a Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania , Philadelphia , PA , USA .,d Ludwig Center for Cancer Research, University of Lausanne , Lausanne , Switzerland
| | - Yadollah Omidi
- a Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania , Philadelphia , PA , USA .,b Research Center for Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
37
|
Jung K, Wang P, Gupta N, Gopal K, Wu F, Ye X, Alshareef A, Bigras G, McMullen TP, Abdulkarim BS, Lai R. Profiling gene promoter occupancy of Sox2 in two phenotypically distinct breast cancer cell subsets using chromatin immunoprecipitation and genome-wide promoter microarrays. Breast Cancer Res 2014; 16:470. [PMID: 25380620 PMCID: PMC4303205 DOI: 10.1186/s13058-014-0470-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 10/20/2014] [Indexed: 11/12/2022] Open
Abstract
Introduction Aberrant expression of the embryonic stem cell marker Sox2 has been reported in breast cancer (BC). We previously identified two phenotypically distinct BC cell subsets separated based on their differential response to a Sox2 transcription activity reporter, namely the reporter-unresponsive (RU) and the more tumorigenic reporter-responsive (RR) cells. We hypothesized that Sox2, as a transcription factor, contributes to their phenotypic differences by mediating differential gene expression in these two cell subsets. Methods We used chromatin immunoprecipitation and a human genome-wide promoter microarray (ChIP-chip) to determine the promoter occupancies of Sox2 in the MCF7 RU and RR breast cancer cell populations. We validated our findings with conventional chromatin immunoprecipitation, quantitative reverse transcription polymerase chain reaction (qPCR), and western blotting using cell lines, and also performed qPCR using patient RU and RR samples. Results We found a largely mutually exclusive profile of gene promoters bound by Sox2 between RU and RR cells derived from MCF7 (1830 and 456 genes, respectively, with only 62 overlapping genes). Sox2 was bound to stem cell- and cancer-associated genes in RR cells. Using quantitative RT-PCR, we confirmed that 15 such genes, including PROM1 (CD133), BMI1, GPR49 (LGR5), and MUC15, were expressed significantly higher in RR cells. Using siRNA knockdown or enforced expression of Sox2, we found that Sox2 directly contributes to the higher expression of these genes in RR cells. Mucin-15, a novel Sox2 downstream target in BC, contributes to the mammosphere formation of BC cells. Parallel findings were observed in the RU and RR cells derived from patient samples. Conclusions In conclusion, our data supports the model that the Sox2 induces differential gene expression in the two distinct cell subsets in BC, and contributes to their phenotypic differences. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0470-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Karen Jung
- Department of Oncology, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada.
| | - Peng Wang
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada.
| | - Nidhi Gupta
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada.
| | - Keshav Gopal
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada.
| | - Fang Wu
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada.
| | - Xiaoxia Ye
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada.
| | - Abdulraheem Alshareef
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada.
| | - Gilbert Bigras
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada.
| | - Todd P McMullen
- Department of Oncology, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada. .,Department of Surgery, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada.
| | | | - Raymond Lai
- Department of Oncology, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada. .,Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada. .,DynaLIFEDx Medical Laboratories, Edmonton, Canada.
| |
Collapse
|
38
|
Cai X, Dai Z, Reeves RS, Caballero-Benitez A, Duran KL, Delrow JJ, Porter PL, Spies T, Groh V. Autonomous stimulation of cancer cell plasticity by the human NKG2D lymphocyte receptor coexpressed with its ligands on cancer cells. PLoS One 2014; 9:e108942. [PMID: 25291178 PMCID: PMC4188595 DOI: 10.1371/journal.pone.0108942] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 08/27/2014] [Indexed: 12/20/2022] Open
Abstract
The stimulatory NKG2D receptor on lymphocytes promotes tumor immune surveillance by targeting ligands selectively induced on cancer cells. Progressing tumors counteract by employing tactics to disable lymphocyte NKG2D. This negative dynamic is escalated as some human cancer cells co-opt expression of NKG2D, thereby complementing the presence of its ligands for autonomous stimulation of oncogenic signaling. Clinical association data imply relationships between cancer cell NKG2D and metastatic disease. Here we show that NKG2D promotes cancer cell plasticity by induction of phenotypic, molecular, and functional signatures diagnostic of the epithelial–mesenchymal transition, and of stem-like traits via induction of Sox9, a key transcriptional regulator of breast stem cell maintenance. These findings obtained with model breast tumor lines and xenotransplants were recapitulated by exvivo cancer cells from primary invasive breast carcinomas. Thus, NKG2D may have the capacity to drive high malignancy traits underlying metastatic disease.
Collapse
Affiliation(s)
- Xin Cai
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Zhenpeng Dai
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Rebecca S. Reeves
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Andrea Caballero-Benitez
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Kate L. Duran
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Jeffrey J. Delrow
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Peggy L. Porter
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Thomas Spies
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- * E-mail:
| | - Veronika Groh
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| |
Collapse
|
39
|
Wu J, Mu Q, Thiviyanathan V, Annapragada A, Vigneswaran N. Cancer stem cells are enriched in Fanconi anemia head and neck squamous cell carcinomas. Int J Oncol 2014; 45:2365-72. [PMID: 25340704 PMCID: PMC4215586 DOI: 10.3892/ijo.2014.2677] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 08/19/2014] [Indexed: 01/06/2023] Open
Abstract
Fanconi anemia (FA) patients have an increased risk of head and neck squamous cell carcinoma (HNSCC) at a higher rate with no apparent risk factors. HNSCC of FA patients is an aggressive tumor characterized by multifocal origin, early metastases and frequent recurrences. Given that cancer stem cells (CSC) drive tumorigenesis, tumor recurrence and metastasis, in this study, we characterized the CSC population in FA and sporadic HNSCC. The Aldefluor assay was used to characterize and isolate CSC with high aldehyde dehydrogenase (ALDH) activity (ALDHpos) in cell lines derived from FA and sporadic HNSCC. Isolated ALDHpos and ALDHneg cells were examined for the expression of stemness genes using reverse transcription-polymerase chain reaction (RT-PCR) array. Tumor cell-derived FA and sporadic HNSCC were examined for their ability to form tumorspheres in vitro. Stem-like cell population in FA and sporadic HNSCC in human and mouse xenograft tumors were evaluated using ALDH isoform 1 (ALDH1) immunohistochemistry. FA‑HNSCC cell lines harbor a greater proportion of ALDHpos cells (15-31%) compared to sporadic HNSCC (10%). Expression of Nanog, Oct-3/4 and Stella, molecular markers of undifferentiated embryonic stem (ES) cells were detected in the ALDHpos FA‑HNSCC cells and not in the ALDHneg cells. FA‑HNSCC cell lines revealed enhanced in vitro tumorsphere formation compared to sporadic HNSCC cells. A higher percentage of ALDH1pos tumor cells are noted in the human and mouse xenograft tumors of FA‑HNSCC compared to sporadic HNSCC tumors. FA‑HNSCC are highly enriched for CSC and may serve as a model to develop CSC-targeted therapies for HNSCC.
Collapse
Affiliation(s)
- Jean Wu
- Department of Diagnostic and Biomedical Sciences, The University of Texas School of Dentistry at Houston, Houston, TX 77054, USA
| | - Qingshan Mu
- Department of Diagnostic and Biomedical Sciences, The University of Texas School of Dentistry at Houston, Houston, TX 77054, USA
| | - Varatharasa Thiviyanathan
- Department of Nanomedicine and Biomedical Engineering, The University of Texas Medical School at Houston, Houston, TX 77030, USA
| | - Ananth Annapragada
- The Singleton Department of Pediatric Radiology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nadarajah Vigneswaran
- Department of Diagnostic and Biomedical Sciences, The University of Texas School of Dentistry at Houston, Houston, TX 77054, USA
| |
Collapse
|
40
|
miR-145 Inhibits Migration and Invasion of Glioma Stem Cells by Targeting ABCG2. Neuromolecular Med 2014; 16:517-28. [DOI: 10.1007/s12017-014-8305-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 04/15/2014] [Indexed: 12/30/2022]
|
41
|
Aloni-Grinstein R, Shetzer Y, Kaufman T, Rotter V. p53: the barrier to cancer stem cell formation. FEBS Lett 2014; 588:2580-9. [PMID: 24560790 DOI: 10.1016/j.febslet.2014.02.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Revised: 02/07/2014] [Accepted: 02/07/2014] [Indexed: 02/08/2023]
Abstract
The role of p53 as the "guardian of the genome" in differentiated somatic cells, triggering various biological processes, is well established. Recent studies in the stem cell field have highlighted a profound role of p53 in stem cell biology as well. These studies, combined with basic data obtained 20 years ago, provide insight into how p53 governs the quantity and quality of various stem cells, ensuring a sufficient repertoire of normal stem cells to enable proper development, tissue regeneration and a cancer free life. In this review we address the role of p53 in genomically stable embryonic stem cells, a unique predisposed cancer stem cell model and adult stem cells, its role in the generation of induced pluripotent stem cells, as well as its role as the barrier to cancer stem cell formation.
Collapse
Affiliation(s)
- Ronit Aloni-Grinstein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yoav Shetzer
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Tom Kaufman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Varda Rotter
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
42
|
Kimura Y, Goi T, Nakazawa T, Hirono Y, Katayama K, Urano T, Yamaguchi A. CD44variant exon 9 plays an important role in colon cancer initiating cells. Oncotarget 2014; 4:785-91. [PMID: 23800986 PMCID: PMC3742838 DOI: 10.18632/oncotarget.1048] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cancer stem cells(cancer initiating cells) have become increasingly important in the treatment of malignant tumors. CD44 in particular has been identified as a marker for stem cells in colon cancer, which is a high-morbidity tumor. However, many details remain unknown, including identification of the relevant exon. The elucidation of these details could lead to the development of new therapies and improvements in prognosis. We report our findings on the importance of CD44 variant exon 9(v9) of stem cells in colon cancer. Using the anti-CD44 standard form(s) antibody, as well as antibodies for each of the CD44 variant exons, we studied colon cancer cell lines by examining stained images of stem cells in the crypt of normal colon mucosa. Using the anti-CD44v9 antibody that fits the normal colon mucosa stem cells, we screened cells using flow cytometry to examine colony formation, resistance to anticancer drugs, and tumor mass formation after subcutaneous implantation in mice. The stem cell–containing region in the crypt of normal colon mucosa was negative for anti-Ki67 antibody staining; only the anti-CD44 v9 antibody stain was expressed. As for colony formation, resistance to anticancer drugs, and tumor mass formation, cells positive both for anti-CD44s and anti-CD44v9 antibody stains was significantly more frequent than those positive for anti-CD44s antibody stain and negative for anti-CD44v9 antibody stain and those negative both for anti-CD44s and anti-CD44v9 antibody stains. CD44 variant exon 9 plays an important role in colon cancer stem cells.
Collapse
Affiliation(s)
- Youhei Kimura
- First Department of Surgery, University of Fukui, Fukui, Japan
| | | | | | | | | | | | | |
Collapse
|
43
|
Garland J. Energy management – a critical role in cancer induction? Crit Rev Oncol Hematol 2013; 88:198-217. [DOI: 10.1016/j.critrevonc.2013.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 03/08/2013] [Accepted: 04/05/2013] [Indexed: 12/18/2022] Open
|
44
|
Osisami M, Keller ET. Mechanisms of Metastatic Tumor Dormancy. J Clin Med 2013; 2:136-50. [PMID: 26237067 PMCID: PMC4470233 DOI: 10.3390/jcm2030136] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 08/20/2013] [Accepted: 09/10/2013] [Indexed: 12/29/2022] Open
Abstract
Tumor metastasis can occur years after an apparent cure due to a phenomenon known as metastatic tumor dormancy; in which tumor masses or individual tumor cells are growth restricted for extended periods of time. This period of dormancy is induced and maintained by several mechanisms, including: (1) Tumor microenvironment factors such as cytokine expression, immunosurveillance and angiogenesis; (2) Metastasis suppressor gene activity; and (3) Cancer therapeutics. Disseminated tumor cells (DTC) are the key cells that result in dormant tumors. However, many challenges exist towards isolating DTCs for mechanistic studies. The main DTC that may represent the dormant cell is the cancer stem cells (CSC) as they have a slow proliferation rate. In addition to limited knowledge regarding induction of tumor dormancy, there are large gaps in knowledge regarding how tumors escape from dormancy. Emerging research into cancer stem cells, immunotherapy, and metastasis suppressor genes, may lead to new approaches for targeted anti-metastatic therapy to prevent dormancy escape. Overall, an enhanced understanding of tumor dormancy is critical for better targeting and treatment of patients to prevent cancer recurrence.
Collapse
Affiliation(s)
- Mary Osisami
- Department of Urology, University of Michigan Medical School, 5111 CCGC1500 E. Medical Center, Ann Arbor, MI 48109-0940, USA.
| | - Evan T Keller
- Department of Urology, University of Michigan Medical School, 5111 CCGC1500 E. Medical Center, Ann Arbor, MI 48109-0940, USA.
| |
Collapse
|
45
|
Abstract
ATP citrate lyase (ACL) knockdown (KD) causes tumor suppression and induces differentiation. We have previously reported that ACL KD reverses epithelial–mesenchymal transition (EMT) in lung cancer cells. Because EMT is often associated with processes that induce stemness, we hypothesized that ACL KD impacts cancer stem cells. By assessing tumorsphere formation and expression of stem cell markers, we showed this to be the case in A549 cells, which harbor a Ras mutation, and in two other non-small-cell lung cancer cell lines, H1975 and H1650, driven by activating EGFR mutations. Inducible ACL KD had the same effect as stable ACL KD. Similar effects were noted in another well-characterized Ras-induced mammary model system (HMLER). Moreover, treatment with hydroxycitrate phenocopied the effects of ACL KD, suggesting that the enzymatic activity of ACL was critical. Indeed, acetate treatment reversed the ACL KD phenotype. Having previously established that ACL KD impacts signaling through the phosphatidylinositol 3-kinase (PI3K) pathway, not the Ras-mitogen-activated protein kinase (MAPK) pathway, and that EMT can be reversed by PI3K inhibitors, we were surprised to find that stemness in these systems was maintained through Ras-MAPK signaling, and not via PI3K signaling. Snail is a downstream transcription factor impacted by Ras-MAPK signaling and known to promote EMT and stemness. We found that snail expression was reduced by ACL KD. In tumorigenic HMLER cells, ACL overexpression increased snail expression and stemness, both of which were reduced by ACL KD. Furthermore, ACL could not initiate either tumorigenesis or stemness by itself. ACL and snail proteins interacted and ACL expression regulated the transcriptional activity of snail. Finally, ACL KD counteracted stem cell characteristics induced in diverse cell systems driven by activation of pathways outside of Ras-MAPK signaling. Our findings unveil a novel aspect of ACL function, namely its impact on cancer stemness in a broad range of genetically diverse cell types.
Collapse
|
46
|
Menendez JA, Joven J, Cufí S, Corominas-Faja B, Oliveras-Ferraros C, Cuyàs E, Martin-Castillo B, López-Bonet E, Alarcón T, Vazquez-Martin A. The Warburg effect version 2.0: metabolic reprogramming of cancer stem cells. Cell Cycle 2013; 12:1166-79. [PMID: 23549172 DOI: 10.4161/cc.24479] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
When fighting cancer, knowledge on metabolism has always been important. Today, it matters more than ever. The restricted cataloging of cancer genomes is quite unlikely to achieve the task of curing cancer, unless it is integrated into metabolic networks that respond to and influence the constantly evolving cancer stem cell (CSC) cellular states. Once the genomic era of carcinogenesis had pushed the 1920s Otto Warburg's metabolic cancer hypothesis into obscurity for decades, the most recent studies begin to support a new developing paradigm, in which the molecular logic behind the conversion of non-CSCs into CSCs can be better understood in terms of the "metabolic facilitators" and "metabolic impediments" that operate as proximate openings and roadblocks, respectively, for the transcriptional events and signal transduction programs that ultimately orchestrate the intrinsic and/or microenvironmental paths to CSC cellular states. Here we propose that a profound understanding of how human carcinomas install a proper "Warburg effect version 2.0" allowing them to "run" the CSCs' "software" programs should guide a new era of metabolo-genomic-personalized cancer medicine. By viewing metabolic reprogramming of CSCs as an essential characteristic that allows dynamic, multidimensional and evolving cancer populations to compete successfully for their expansion on the organism, we now argue that CSCs bioenergetics might be another cancer hallmark. A definitive understanding of metabolic reprogramming in CSCs may complement or to some extent replace, the 30-y-old paradigm of targeting oncogenes to treat human carcinomas, because it can be possible to metabolically create non-permissive or "hostile" metabotypes to prevent the occurrence of CSC cellular states with tumor- and metastasis-initiating capacity.
Collapse
Affiliation(s)
- Javier A Menendez
- Metabolism & Cancer Group, Translational Research Laboratory, Catalan Institute of Oncology-Girona (ICO-Girona), Girona, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lee HR, Kim MJ, Ha G, Kim SJ, Kim SH, Kang CD. Presence of Leukemia-maintaining Cells in Differentiation-resistant Fraction of K562 Chronic Myelogenous Leukemia. ACTA ACUST UNITED AC 2013. [DOI: 10.5352/jls.2013.23.2.197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
48
|
Yi SY, Hao YB, Nan KJ, Fan TL. Cancer stem cells niche: a target for novel cancer therapeutics. Cancer Treat Rev 2012; 39:290-6. [PMID: 23219150 DOI: 10.1016/j.ctrv.2012.10.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 10/05/2012] [Accepted: 10/08/2012] [Indexed: 12/18/2022]
Abstract
Nowadays, cancer has been a frequent disease, and the first or second most common cause of death worldwide. Despite a better understanding of the biology of cancer cells, the therapy of most cancers has not significantly changed for the past four decades. It is because conventional chemotherapies and/or radiation therapies are usually designed to eradicate highly proliferative cells. Mounting evidence has implicated that cancer is a disease of stem cells. Cancer stem cells (CSC) are often relatively quiescent, and therefore may not be affected by therapies targeting rapidly dividing cells. Like normal stem cells (NSC) residing in a "stem cell niche" that maintains them in a stem-like state, CSC also require a special microenvironment to control their self-renewal and undifferentiated state. The "CSC niche" is likely to be the most crucial target in the treatment of cancer. In this article, we summarize the current knowledge regarding CSC and their niche microenvironments. Understanding of CSC's origin, molecular profile, and interaction with their microenvironments, this could be a paradigm shift in the treatment of cancer, away from targeting the blast cells and towards the targeting of the CSC, thus improving therapeutic outcome.
Collapse
Affiliation(s)
- Shan-Yong Yi
- Department of Oncology of the Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zheng Zhou 450007, Henan Province, China.
| | | | | | | |
Collapse
|
49
|
Vira D, Basak SK, Veena MS, Wang MB, Batra RK, Srivatsan ES. Cancer stem cells, microRNAs, and therapeutic strategies including natural products. Cancer Metastasis Rev 2012; 31:733-51. [PMID: 22752409 DOI: 10.1007/s10555-012-9382-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Embryonic stem cells divide continuously and differentiate into organs through the expression of specific transcription factors at specific time periods. Differentiated adult stem cells on the other hand remain in quiescent state and divide by receiving cues from the environment (extracellular matrix or niche), as in the case of wound healing from tissue injury or inflammation. Similarly, it is believed that cancer stem cells (CSCs), forming a smaller fraction of the tumor bulk, also remain in a quiescent state. These cells are capable of initiating and propagating neoplastic growth upon receiving environmental cues, such as overexpression of growth factors, cytokines, and chemokines. Candidate CSCs express distinct biomarkers that can be utilized for their identification and isolation. This review focuses on the known and candidate cancer stem cell markers identified in various solid tumors and the promising future of disease management and therapy targeted at these markers. The review also provides details on the differential expression of microRNAs (miRNAs), and the miRNA- and natural product-based therapies that could be applied for the treatment of cancer stem cells.
Collapse
Affiliation(s)
- Darshni Vira
- Department of Surgery, VAGLAHS West Los Angeles, Los Angeles, CA 90073, USA
| | | | | | | | | | | |
Collapse
|
50
|
Oishi N, Kumar MR, Roessler S, Ji J, Forgues M, Budhu A, Zhao X, Andersen JB, Ye QH, Jia HL, Qin LX, Yamashita T, Woo HG, Kim YJ, Kaneko S, Tang ZY, Thorgeirsson SS, Wang XW. Transcriptomic profiling reveals hepatic stem-like gene signatures and interplay of miR-200c and epithelial-mesenchymal transition in intrahepatic cholangiocarcinoma. Hepatology 2012; 56:1792-803. [PMID: 22707408 PMCID: PMC3458130 DOI: 10.1002/hep.25890] [Citation(s) in RCA: 181] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 05/19/2012] [Indexed: 12/14/2022]
Abstract
UNLABELLED Intrahepatic cholangiocellular carcinoma (ICC) is the second most common type of primary liver cancer. However, its tumor heterogeneity and molecular characteristics are largely unknown. In this study, we conducted transcriptomic profiling of 23 ICC and combined hepatocellular cholangiocarcinoma tumor specimens from Asian patients using Affymetrix messenger RNA (mRNA) and NanoString microRNA microarrays to search for unique gene signatures linked to tumor subtypes and patient prognosis. We validated the signatures in an additional 68 ICC cases derived from Caucasian patients. We found that both mRNA and microRNA expression profiles could independently classify Asian ICC cases into two main subgroups, one of which shared gene expression signatures with previously identified hepatocellular carcinoma (HCC) with stem cell gene expression traits. ICC-specific gene signatures could predict survival in Asian HCC cases and independently in Caucasian ICC cases. Integrative analyses of the ICC-specific mRNA and microRNA expression profiles revealed that a common signaling pathway linking miR-200c signaling to epithelial-mesenchymal transition (EMT) was preferentially activated in ICC with stem cell gene expression traits. Inactivation of miR-200c resulted in an induction of EMT, whereas activation of miR-200c led to a reduction of EMT including a reduced cell migration and invasion in ICC cells. We also found that miR-200c and neural cell adhesion molecule 1 (NCAM1) expression were negatively correlated and their expression levels were predictive of survival in ICC samples. NCAM1, a known hepatic stem/progenitor cell marker, was experimentally demonstrated to be a direct target of miR-200c. CONCLUSION Our results indicate that ICC and HCC share common stem-like molecular characteristics and poor prognosis. We suggest that the specific components of EMT may be exploited as critical biomarkers and clinically relevant therapeutic targets for an aggressive form of stem cell-like ICC.
Collapse
Affiliation(s)
- Naoki Oishi
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mia R. Kumar
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stephanie Roessler
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Junfang Ji
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marshonna Forgues
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anuradha Budhu
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xuelian Zhao
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jesper B. Andersen
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Qing-Hai Ye
- Liver Cancer Institute, Fudan University, Shanghai, China
| | - Hu-Liang Jia
- Liver Cancer Institute, Fudan University, Shanghai, China
| | - Lun-Xiu Qin
- Liver Cancer Institute, Fudan University, Shanghai, China
| | - Taro Yamashita
- Liver Disease Center and Kanazawa University Hospital, Kanazawa University, Kanazawa, Japan
| | - Hyun Goo Woo
- Department of Physiology, Ajou University School of Medicine, Suwon
| | - Yoon Jun Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Shuichi Kaneko
- Liver Disease Center and Kanazawa University Hospital, Kanazawa University, Kanazawa, Japan
| | - Zhao-You Tang
- Liver Cancer Institute, Fudan University, Shanghai, China
| | - Snorri S. Thorgeirsson
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA,Address reprint request to: Xin Wei Wang, National Cancer Institute, 37 Convent Drive, Building 37, Room 3044A, Bethesda, MD 20892;
| |
Collapse
|