1
|
Cruz SM, Iranpur KR, Judge SJ, Ames E, Sturgill IR, Farley LE, Darrow MA, Crowley JS, Monjazeb AM, Murphy WJ, Canter RJ. Low-Dose Sorafenib Promotes Cancer Stem Cell Expansion and Accelerated Tumor Progression in Soft Tissue Sarcomas. Int J Mol Sci 2024; 25:3351. [PMID: 38542325 PMCID: PMC10969893 DOI: 10.3390/ijms25063351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 08/03/2024] Open
Abstract
The cancer stem cell (CSC) hypothesis postulates that heterogeneous human cancers harbor a population of stem-like cells which are resistant to cytotoxic therapies, thus providing a reservoir of relapse following conventional therapies like chemotherapy and radiation (RT). CSCs have been observed in multiple human cancers, and their presence has been correlated with worse clinical outcomes. Here, we sought to evaluate the impact of drug dosing of the multi-tyrosine kinase inhibitor, sorafenib, on CSC and non-CSCs in soft tissue sarcoma (STS) models, hypothesizing differential effects of sorafenib based on dose and target cell population. In vitro, human cancer cell lines and primary STS from surgical specimens were exposed to escalating doses of sorafenib to determine cell viability and expression of CSC marker aldehyde dehydrogenase (ALDH). In vivo, ALDHbright CSCs were isolated, exposed to sorafenib, and xenograft growth and survival analyses were performed. We observed that sarcoma CSCs appear to paradoxically respond to the tyrosine kinase inhibitor sorafenib at low doses with increased proliferation and stem-like function of CSCs, whereas anti-viability effects dominated at higher doses. Importantly, STS patients receiving neoadjuvant sorafenib and RT on a clinical trial (NCT00864032) showed increased CSCs post therapy, and higher ALDH scores post therapy were associated with worse metastasis-free survival. These data suggest that low-dose sorafenib may promote the CSC phenotype in STS with clinically significant effects, including increased tumor growth and higher rates of metastasis formation in sarcoma patients.
Collapse
Affiliation(s)
- Sylvia M. Cruz
- Division of Surgical Oncology, Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
| | - Khurshid R. Iranpur
- Division of Surgical Oncology, Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
| | - Sean J. Judge
- Division of Surgical Oncology, Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
| | - Erik Ames
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Ian R. Sturgill
- Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lauren E. Farley
- Division of Surgical Oncology, Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
| | - Morgan A. Darrow
- Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Jiwon Sarah Crowley
- Division of Surgical Oncology, Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
| | - Arta M. Monjazeb
- Department of Radiation Oncology, University of California Davis, Sacramento, CA 95817, USA
| | - William J. Murphy
- Department of Dermatology, University of California Davis, Sacramento, CA 95817, USA;
| | - Robert J. Canter
- Division of Surgical Oncology, Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
| |
Collapse
|
2
|
Pavlova G, Kolesnikova V, Samoylenkova N, Drozd S, Revishchin A, Shamadykova D, Usachev DY, Kopylov A. A Combined Effect of G-Quadruplex and Neuro-Inducers as an Alternative Approach to Human Glioblastoma Therapy. Front Oncol 2022; 12:880740. [PMID: 35586496 PMCID: PMC9109612 DOI: 10.3389/fonc.2022.880740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/28/2022] [Indexed: 11/22/2022] Open
Abstract
Cancer cell reprogramming based on treatment with G-quadruplex, having antiproliferative power, along with small molecules able to develop iPSCs into neurons, could create a novel approach to diminish the chance of glioblastoma recurrence and circumvent tumor resistance to conventional therapy. In this research, we have tested several combinations of factors to affect both total cell cultures, derived from tumor tissue of patients after surgical resection and two subfractions of this cell culture after dividing them into CD133-enriched and CD133-depleted populations (assuming CD133 to be a marker of glioblastoma stem-like cells). CD133+ and CD133− cells exhibit different responses to the same combinations of factors; CD133+ cells have stem-like properties and are more resistant. Therefore, the ability to affect CD133+ cells provides a possibility to circumvent resistance to conventional therapy and to build a promising strategy for translation to improve the treatment of patients with glioblastoma.
Collapse
Affiliation(s)
- Galina Pavlova
- Laboratory of Neurogenetics and Genetics Development, Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences (RAS), Moscow, Russia.,Federal State Autonomous Institution «N. N. Burdenko National Medical Research Center of Neurosurgery» of the Ministry of Health of the Russian Federation, Moscow, Russia.,Department of Medical Genetics, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Varvara Kolesnikova
- Laboratory of Neurogenetics and Genetics Development, Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences (RAS), Moscow, Russia
| | - Nadezhda Samoylenkova
- Federal State Autonomous Institution «N. N. Burdenko National Medical Research Center of Neurosurgery» of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Sergey Drozd
- Federal State Autonomous Institution «N. N. Burdenko National Medical Research Center of Neurosurgery» of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alexander Revishchin
- Laboratory of Neurogenetics and Genetics Development, Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences (RAS), Moscow, Russia
| | - Dzhirgala Shamadykova
- Laboratory of Neurogenetics and Genetics Development, Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences (RAS), Moscow, Russia
| | - Dmitry Y Usachev
- Federal State Autonomous Institution «N. N. Burdenko National Medical Research Center of Neurosurgery» of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alexey Kopylov
- Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
3
|
Koltai T, Reshkin SJ, Carvalho TMA, Di Molfetta D, Greco MR, Alfarouk KO, Cardone RA. Resistance to Gemcitabine in Pancreatic Ductal Adenocarcinoma: A Physiopathologic and Pharmacologic Review. Cancers (Basel) 2022; 14:2486. [PMID: 35626089 PMCID: PMC9139729 DOI: 10.3390/cancers14102486] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a very aggressive tumor with a poor prognosis and inadequate response to treatment. Many factors contribute to this therapeutic failure: lack of symptoms until the tumor reaches an advanced stage, leading to late diagnosis; early lymphatic and hematic spread; advanced age of patients; important development of a pro-tumoral and hyperfibrotic stroma; high genetic and metabolic heterogeneity; poor vascular supply; a highly acidic matrix; extreme hypoxia; and early development of resistance to the available therapeutic options. In most cases, the disease is silent for a long time, andwhen it does become symptomatic, it is too late for ablative surgery; this is one of the major reasons explaining the short survival associated with the disease. Even when surgery is possible, relapsesare frequent, andthe causes of this devastating picture are the low efficacy ofand early resistance to all known chemotherapeutic treatments. Thus, it is imperative to analyze the roots of this resistance in order to improve the benefits of therapy. PDAC chemoresistance is the final product of different, but to some extent, interconnected factors. Surgery, being the most adequate treatment for pancreatic cancer and the only one that in a few selected cases can achieve longer survival, is only possible in less than 20% of patients. Thus, the treatment burden relies on chemotherapy in mostcases. While the FOLFIRINOX scheme has a slightly longer overall survival, it also produces many more adverse eventsso that gemcitabine is still considered the first choice for treatment, especially in combination with other compounds/agents. This review discusses the multiple causes of gemcitabine resistance in PDAC.
Collapse
Affiliation(s)
| | - Stephan Joel Reshkin
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (D.D.M.); (M.R.G.); (R.A.C.)
| | - Tiago M. A. Carvalho
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (D.D.M.); (M.R.G.); (R.A.C.)
| | - Daria Di Molfetta
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (D.D.M.); (M.R.G.); (R.A.C.)
| | - Maria Raffaella Greco
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (D.D.M.); (M.R.G.); (R.A.C.)
| | - Khalid Omer Alfarouk
- Zamzam Research Center, Zamzam University College, Khartoum 11123, Sudan;
- Alfarouk Biomedical Research LLC, Temple Terrace, FL 33617, USA
| | - Rosa Angela Cardone
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (D.D.M.); (M.R.G.); (R.A.C.)
| |
Collapse
|
4
|
Shiozaki A, Konishi T, Kosuga T, Kudou M, Kurashima K, Inoue H, Shoda K, Arita T, Konishi H, Morimura R, Komatsu S, Ikoma H, Toma A, Kubota T, Fujiwara H, Okamoto K, Otsuji E. Roles of voltage‑gated potassium channels in the maintenance of pancreatic cancer stem cells. Int J Oncol 2021; 59:76. [PMID: 34414448 PMCID: PMC8425586 DOI: 10.3892/ijo.2021.5256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 06/16/2021] [Indexed: 11/10/2022] Open
Abstract
The targeting of membrane proteins that are activated in cancer stem cells (CSCs) represents one of the key recent strategies in cancer therapy. The present study analyzed ion channel expression profiles and functions in pancreatic CSCs (PCSCs). Cells strongly expressing aldehyde dehydrogenase 1 family member A1 (ALDH1A1) were isolated from the human pancreatic PK59 cell line using fluorescence-activated cell sorting, and PCSCs were identified based on tumorsphere formation. Microarray analysis was performed to investigate the gene expression profiles in PCSCs. ALDH1A1 messenger RNA levels were higher in PCSCs compared with non-PCSCs. PCSCs were resistant to 5-fluorouracil and capable of redifferentiation. The results of the microarray analysis revealed that gene expression related to ion channels, including voltage-gated potassium channels (Kv), was upregulated in PCSCs compared with non-PCSCs. 4-Aminopyridine (4-AP), a potent Kv inhibitor, exhibited greater cytotoxicity in PCSCs compared with non-PCSCs. In a xenograft model in nude mice, tumor volumes were significantly lower in mice inoculated with PK59 cells pre-treated with 4-AP compared with those in mice injected with non-treated cells. The present results identified a role of Kv in the persistence of PCSCs and suggested that the Kv inhibitor 4-AP may have potential as a therapeutic agent for pancreatic carcinoma.
Collapse
Affiliation(s)
- Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Tomoki Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Toshiyuki Kosuga
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Michihiro Kudou
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Kento Kurashima
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Hiroyuki Inoue
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Katsutoshi Shoda
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Tomohiro Arita
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Ryo Morimura
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Hisashi Ikoma
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Atsushi Toma
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Takeshi Kubota
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Hitoshi Fujiwara
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Kazuma Okamoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| |
Collapse
|
5
|
de Maria S, Santoro A, Fuggetta MP, Rocchetti R, Cottarelli A, Lanzilli G, Stiuso P, Angelico G, Spadola S, Franco Zannoni G, Rubini C, Emanuelli M, Carmela Pedicillo M, Pannone G, Muzio LL. A possible interplay between HR-HPV and stemness in tumor development: an in vivo investigation of CD133 as a putative marker of cancer stem cell in HPV18-infected KB cell line. APMIS 2020; 128:637-646. [PMID: 32911563 DOI: 10.1111/apm.13078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 09/08/2020] [Indexed: 12/30/2022]
Abstract
High-risk HPVs (HR-HPVs) are DNA viruses considered as primary etiologic factors in malignancies of the low female genital tract. Their presence has also been documented in oropharyngeal and laryngeal cancers. However, HPV infection is considered a necessary but not sufficient cause of tumoral development; meantime, increasing evidences on the tumorigenic role of cancer stem cells (CSCs) have been documented in the literature. CSCs represent a small subpopulation of neoplastic cells with self-renewal potential, capable of maintaining tumor growth and cell differentiation, also involved in metastatic process, recurrence, and resistance to chemotherapeutic agents. In the present study, performed on KB cell lines, we evaluated the tumor forming potential of CSCs, and their relationship with the HPV infection status. We started our study by identifying the most aggressive cell line on the minimal number of cells being able of growth in vivo in a model of athymic nude mice (BALB/c nu/nu). We used an oral-derived KB cell line separated in the KB-CD133+ and KB-CD133- populations, by using immunomagnetic beads and fluorescence-activated cell sorting (FACS). The separated populations were injected in athymic nude mice (BALB/c nu/nu). Xenograft tumors have been analyzed for tumor size, CD133 expression by immunohistochemistry (IHC) and for DNA HR-HPV integration by in situ hybridization (ISH), comparing CD133-enriched xenograft tumors versus the CD133 non-enriched ones. On standard conditions, the KB cell line has a poor population of glycosylated CD133 marker (<5.0%) when investigated with antibodies versus CD133, and more specifically its glycosylated epitope (AC133). Enriched CD133 KB cells possess a higher capacity of tumor growth in xenograft models of nude mice when compared to KB CD133-negative cells. We observed that the AC133 epitope, extensively used to purifying hematopoietic stem cells, is able to select an epithelial subpopulation of cancer stem cells with aggressive behavior. We retain that CD133 may be a useful target in anticancer strategies including pharmacological and immunological therapies.
Collapse
Affiliation(s)
- Salvatore de Maria
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy.,GLURES, Academic SPINOFF Ca Foscari University of Venice, Venice, Italy
| | - Angela Santoro
- Unità di Gineco-Patologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanita Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Maria Pia Fuggetta
- GLURES, Academic SPINOFF Ca Foscari University of Venice, Venice, Italy.,Istituto di Farmacologia Traslazionale, Area Ricerca Tor Vergata, CNR, Rome, Italy
| | - Romina Rocchetti
- Dipartimento di Neuroscienze, Sezione di Anatomia Patologica, Università Politecnica delle Marche, Ancona, Italy
| | - Andrea Cottarelli
- Istituto di Farmacologia Traslazionale, Area Ricerca Tor Vergata, CNR, Rome, Italy
| | - Giulia Lanzilli
- Istituto di Farmacologia Traslazionale, Area Ricerca Tor Vergata, CNR, Rome, Italy
| | - Paola Stiuso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giuseppe Angelico
- Unità di Gineco-Patologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanita Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Saveria Spadola
- Unità di Gineco-Patologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanita Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Gian Franco Zannoni
- Unità di Gineco-Patologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanita Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy.,Istituto di Anatomia Patologica, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Corrado Rubini
- Dipartimento di Neuroscienze, Sezione di Anatomia Patologica, Università Politecnica delle Marche, Ancona, Italy
| | - Monica Emanuelli
- Dipartimento di Neuroscienze, Sezione di Anatomia Patologica, Università Politecnica delle Marche, Ancona, Italy
| | | | - Giuseppe Pannone
- Dipartimento di Scienze Chirurgiche, Università degli Studi di Foggia, Foggia, Italy
| | - Lorenzo Lo Muzio
- Dipartimento di Scienze Chirurgiche, Università degli Studi di Foggia, Foggia, Italy
| |
Collapse
|
6
|
Ambrosini G, Dalla Pozza E, Fanelli G, Di Carlo C, Vettori A, Cannino G, Cavallini C, Carmona-Carmona CA, Brandi J, Rinalducci S, Scupoli MT, Rasola A, Cecconi D, Palmieri M, Dando I. Progressively De-Differentiated Pancreatic Cancer Cells Shift from Glycolysis to Oxidative Metabolism and Gain a Quiescent Stem State. Cells 2020; 9:cells9071572. [PMID: 32605166 PMCID: PMC7408749 DOI: 10.3390/cells9071572] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is typically characterized by high chemoresistance and metastatic spread, features mainly attributable to cancer stem cells (CSCs). It is of central interest the characterization of CSCs and, in particular, the study of their metabolic features in order to selectively identify their peculiarities for an efficient therapeutic approach. In this study, CSCs have been obtained by culturing different PDAC cell lines with a specific growth medium. Cells were characterized for the typical stem/mesenchymal properties at short-, medium-, and long-term culture. Metabolomics, proteomics, analysis of oxygen consumption rate in live cells, and the effect of the inhibition of lactate transporter on cell proliferation have been performed to delineate the metabolism of CSCs. We show that gradually de-differentiated pancreatic cancer cells progressively increase the expression of both stem and epithelial-to-mesenchymal transition markers, shift their metabolism from a glycolytic to an oxidative one, and lastly gain a quiescent state. These quiescent stem cells are characterized by high chemo-resistance, clonogenic ability, and metastatic potential. Re-differentiation reverts these features, re-activating their proliferative capacity and glycolytic metabolism, which generally correlates with high aggressiveness. These observations add an important piece of knowledge to the comprehension of the biology of CSCs, whose metabolic plasticity could be exploited for the generation of promising and selective therapeutic approaches for PDAC patients.
Collapse
Affiliation(s)
- Giulia Ambrosini
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (G.A.); (E.D.P.); (C.A.C.-C.); (M.T.S.); (M.P.)
| | - Elisa Dalla Pozza
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (G.A.); (E.D.P.); (C.A.C.-C.); (M.T.S.); (M.P.)
| | - Giuseppina Fanelli
- Department of Ecological and Biological Sciences, University of Tuscia, 01100 Viterbo, Italy; (G.F.); (S.R.)
| | - Claudia Di Carlo
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (C.D.C.); (A.V.); (J.B.); (D.C.)
| | - Andrea Vettori
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (C.D.C.); (A.V.); (J.B.); (D.C.)
| | - Giuseppe Cannino
- Department of Biomedical Sciences, University of Padova, 35122 Padova, Italy; (G.C.); (A.R.)
| | - Chiara Cavallini
- Research Center LURM (Interdepartmental Laboratory of Medical Research), University of Verona, 37134 Verona, Italy;
| | - Cristian Andres Carmona-Carmona
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (G.A.); (E.D.P.); (C.A.C.-C.); (M.T.S.); (M.P.)
| | - Jessica Brandi
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (C.D.C.); (A.V.); (J.B.); (D.C.)
| | - Sara Rinalducci
- Department of Ecological and Biological Sciences, University of Tuscia, 01100 Viterbo, Italy; (G.F.); (S.R.)
| | - Maria Teresa Scupoli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (G.A.); (E.D.P.); (C.A.C.-C.); (M.T.S.); (M.P.)
- Research Center LURM (Interdepartmental Laboratory of Medical Research), University of Verona, 37134 Verona, Italy;
| | - Andrea Rasola
- Department of Biomedical Sciences, University of Padova, 35122 Padova, Italy; (G.C.); (A.R.)
| | - Daniela Cecconi
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (C.D.C.); (A.V.); (J.B.); (D.C.)
| | - Marta Palmieri
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (G.A.); (E.D.P.); (C.A.C.-C.); (M.T.S.); (M.P.)
| | - Ilaria Dando
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (G.A.); (E.D.P.); (C.A.C.-C.); (M.T.S.); (M.P.)
- Correspondence: ; Tel.: +39-045-8027174
| |
Collapse
|
7
|
Prasad S, Ramachandran S, Gupta N, Kaushik I, Srivastava SK. Cancer cells stemness: A doorstep to targeted therapy. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165424. [PMID: 30818002 DOI: 10.1016/j.bbadis.2019.02.019] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/15/2019] [Accepted: 02/20/2019] [Indexed: 02/07/2023]
Abstract
Recent advances in research on cancer have led to understand the pathogenesis of cancer and development of new anticancer drugs. Despite of these advancements, many tumors have been found to recur, undergo metastasis and develop resistance to therapy. Accumulated evidences suggest that small population of cancer cells known as cancer stem cells (CSC) are responsible for reconstitution and propagation of the disease. CSCs possess the ability to self-renew, differentiate and proliferate like normal stem cells. CSCs also appear to have resistance to anti-cancer therapies and subsequent relapse. The underlying stemness properties of the CSCs are reliant on multiple molecular targets such as signaling pathways, cell surface molecules, tumor microenvironment, apoptotic pathways, microRNA, stem cell differentiation, and drug resistance markers. Thus an effective therapeutic strategy relies on targeting CSCs to overcome the possible tumor relapse and chemoresistance. The targeted inhibition of these stem cell biomarkers is one of the promising approaches to eliminate cancer stemness. This review article summarizes possible targets of cancer cell stemness for the complete treatment of cancer.
Collapse
Affiliation(s)
- Sahdeo Prasad
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Sharavan Ramachandran
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Nehal Gupta
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Itishree Kaushik
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Sanjay K Srivastava
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA.
| |
Collapse
|
8
|
Cancer stem cell fate determination: a nuclear phenomenon. THE NUCLEUS 2019. [DOI: 10.1007/s13237-019-00281-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
9
|
p21-Activated kinase 3 promotes cancer stem cell phenotypes through activating the Akt-GSK3β-β-catenin signaling pathway in pancreatic cancer cells. Cancer Lett 2019; 456:13-22. [PMID: 31051214 DOI: 10.1016/j.canlet.2019.04.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 04/18/2019] [Accepted: 04/25/2019] [Indexed: 12/18/2022]
|
10
|
Dang SC, Qian XB, Jin W, Cui L, Chen JX, Gu M. G-protein-signaling modulator 2 expression and role in a CD133 + pancreatic cancer stem cell subset. Onco Targets Ther 2019; 12:785-794. [PMID: 30774366 PMCID: PMC6352860 DOI: 10.2147/ott.s187670] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND To investigate the expression and role of G-protein-signaling modulator 2 (GPSM2) in a CD133+ pancreatic stem cell subset. MATERIALS AND METHODS Pancreatic cancer stem cells (PCSCs) from the cell line PANC-1 were sorted into CD133+ and CD133- subsets by flow cytometry. The tumorigenic potential of the subsets was assessed by subcutaneous tumor formation experiments in nude mice. Differential expression of GPSM2 was examined by real-time quantitative-PCR (qPCR) and Western blotting. To silence GPSM2 expression, a shRNA lentiviral vector targeting GPSM2 was constructed and stably transfected into CD133+ PCSCs. The inhibitory efficiency of the GPSM2 gene was verified by qPCR and Western blotting. The proliferation, colony formation, and migration abilities of the transfected CD133+ pancreatic cancer cells were assessed by MTT, soft agar colony formation, and Transwell assays. RESULTS CD133+ and CD133- cell subsets were successfully isolated from PANC-1 cells. The CD133+ subset subcutaneously formed tumors in nude mice that were significantly bigger (343.05±57.59 mm3 vs 176.86±32.58 mm3, P<0.01) and denser (4.13±0.37 g vs 1.07±0.21 g, P<0.01) than those of the CD133- group. The GPSM2 mRNA and protein expression was significantly higher in CD133+ cells than in CD133- cells. Stable downregulation of GPSM2 expression reduced the proliferation, colony formation, and migration abilities of CD133+ PANC-1 cells (P<0.05). CONCLUSION The CD133+PANC-1 cells have obvious stem cell characteristics and increased GPSM2 expression. Downregulation of GPSM2 significantly reduces the proliferation and migration ability of the cells. Therefore, GPSM2 may provide an important target for regulating PCSCs.
Collapse
Affiliation(s)
- Sheng-Chun Dang
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, People's Republic of China
| | - Xiao-Bao Qian
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, People's Republic of China
| | - Wei Jin
- Department of Obstetrics and Gynecology, ChangShu No. 2 People's Hospital, Changshu, Jiangsu 215500, People's Republic of China
| | - Lei Cui
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, People's Republic of China
| | - Ji-Xiang Chen
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, People's Republic of China
| | - Min Gu
- Department of Oncology, Zhenjiang Hospital of Traditional Chinese and Western Medicine, Zhenjiang, Jiangsu 212001, People's Republic of China,
| |
Collapse
|
11
|
Jadaun A, Sharma S, Verma R, Dixit A. Pinostrobin inhibits proliferation and induces apoptosis in cancer stem-like cells through a reactive oxygen species-dependent mechanism. RSC Adv 2019; 9:12097-12109. [PMID: 35516989 PMCID: PMC9063484 DOI: 10.1039/c8ra08380k] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 03/19/2019] [Indexed: 12/30/2022] Open
Abstract
Current treatments and targeted therapies for malignancies are limited due to their severe toxicity and the development of resistance against such treatments, which leads to relapse. Past evidence has indicated that a number of plant-derived dietary agents possess biological activity against highly tumorigenic and resistant cell populations associated with cancer relapse. These subpopulations, termed cancer stem-like cells (CSCs), have been targeted with plant-derived dietary flavonoids. The present study was undertaken to assess the anti-proliferative potential of pinostrobin, a dietary flavonoid, against CSCs. Sphere-forming cells were developed from HeLa cell lines using specific culture conditions. The existence of a CSC population was confirmed by the morphological examination and analysis of surface markers using confocal microscopy and flow cytometry. The effect of pinostrobin on the cell viability of the CSC population, evaluated through MTT reduction assays and the expression levels of surface markers (CD44+ and CD24+), was studied through various biological assays. HeLa-derived CSCs showed higher CD44+ and lower CD24+ expression. Pinostrobin inhibited the self-renewal capacity and sphere formation efficiency of CSCs in a dose-dependent manner. Increased ROS production, and decreased mitochondrial membrane potential and CD44+ expression indicated that pinostrobin promoted ROS-mediated apoptosis in CSCs. These results thus demonstrate the therapeutic potential and effectiveness of pinostrobin in the chemoprevention and relapse of cancer by targeting the CSC population. Thus, pinostrobin, in combination with currently available chemo and radiation therapies, could possibly be used as a safe strategy to alleviate adverse treatment effects, together with enhancing the efficacy. The anti-proliferative potential of pinostrobin, a dietary flavonoid, is evaluated against cancer stem-like cells.![]()
Collapse
Affiliation(s)
- Alka Jadaun
- Gene Regulation Laboratory
- School of Biotechnology
- Jawaharlal Nehru University
- New Delhi-110067
- India
| | - Sapna Sharma
- Gene Regulation Laboratory
- School of Biotechnology
- Jawaharlal Nehru University
- New Delhi-110067
- India
| | - Radha Verma
- Gene Regulation Laboratory
- School of Biotechnology
- Jawaharlal Nehru University
- New Delhi-110067
- India
| | - Aparna Dixit
- Gene Regulation Laboratory
- School of Biotechnology
- Jawaharlal Nehru University
- New Delhi-110067
- India
| |
Collapse
|
12
|
Liu L, Zhi Q, Shen M, Gong FR, Zhou BP, Lian L, Shen B, Chen K, Duan W, Wu MY, Tao M, Li W. FH535, a β-catenin pathway inhibitor, represses pancreatic cancer xenograft growth and angiogenesis. Oncotarget 2018; 7:47145-47162. [PMID: 27323403 PMCID: PMC5216931 DOI: 10.18632/oncotarget.9975] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 05/17/2016] [Indexed: 12/30/2022] Open
Abstract
The WNT/β-catenin pathway plays an important role in pancreatic cancer carcinogenesis. We evaluated the correlation between aberrant β-catenin pathway activation and the prognosis pancreatic cancer, and the potential of applying the β-catenin pathway inhibitor FH535 to pancreatic cancer treatment. Meta-analysis and immunohistochemistry showed that abnormal β-catenin pathway activation was associated with unfavorable outcome. FH535 repressed pancreatic cancer xenograft growth in vivo. Gene Ontology (GO) analysis of microarray data indicated that target genes responding to FH535 participated in stemness maintenance. Real-time PCR and flow cytometry confirmed that FH535 downregulated CD24 and CD44, pancreatic cancer stem cell (CSC) markers, suggesting FH535 impairs pancreatic CSC stemness. GO analysis of β-catenin chromatin immunoprecipitation sequencing data identified angiogenesis-related gene regulation. Immunohistochemistry showed that higher microvessel density correlated with elevated nuclear β-catenin expression and unfavorable outcome. FH535 repressed the secretion of the proangiogenic cytokines vascular endothelial growth factor (VEGF), interleukin (IL)-6, IL-8, and tumor necrosis factor-α, and also inhibited angiogenesis in vitro and in vivo. Protein and mRNA microarrays revealed that FH535 downregulated the proangiogenic genes ANGPT2, VEGFR3, IFN-γ, PLAUR, THPO, TIMP1, and VEGF. FH535 not only represses pancreatic CSC stemness in vitro, but also remodels the tumor microenvironment by repressing angiogenesis, warranting further clinical investigation.
Collapse
Affiliation(s)
- Lu Liu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiaoming Zhi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng Shen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fei-Ran Gong
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Binhua P Zhou
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA.,Departments of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Lian Lian
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Oncology, Suzhou Xiangcheng People's Hospital, Suzhou, China.,Department of Pathology, Suzhou Xiangcheng People's Hospital, Suzhou, China
| | - Bairong Shen
- Center for Systems Biology, Soochow University, Suzhou, China
| | - Kai Chen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Weiming Duan
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng-Yao Wu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Tao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China.,PREMED Key Laboratory for Precision Medicine, Soochow University, Suzhou, China.,Jiangsu Institute of Clinical Immunology, Suzhou, China.,Institute of Medical Biotechnology, Soochow University, Suzhou, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Markey Cancer Center, University of Kentucky, Lexington, KY, USA.,Center for Systems Biology, Soochow University, Suzhou, China.,PREMED Key Laboratory for Precision Medicine, Soochow University, Suzhou, China.,Jiangsu Institute of Clinical Immunology, Suzhou, China
| |
Collapse
|
13
|
Giovannetti E, van der Borden CL, Frampton AE, Ali A, Firuzi O, Peters GJ. Never let it go: Stopping key mechanisms underlying metastasis to fight pancreatic cancer. Semin Cancer Biol 2017; 44:43-59. [PMID: 28438662 DOI: 10.1016/j.semcancer.2017.04.006] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/12/2017] [Accepted: 04/18/2017] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive neoplasm, predicted to become the second leading cause of cancer-related deaths before 2030. This dismal trend is mainly due to lack of effective treatments against its metastatic behavior. Therefore, a better understanding of the key mechanisms underlying metastasis should provide new opportunities for therapeutic purposes. Genomic analyses revealed that aberrations that fuel PDAC tumorigenesis and progression, such as SMAD4 loss, are also implicated in metastasis. Recently, microRNAs have been shown to play a regulatory role in the metastatic behavior of many tumors, including PDAC. In particular, miR-10 and miR-21 have appeared as master regulators of the metastatic program, while members of the miR-200 family are involved in the epithelial-to-mesenchymal switch, favoring cell migration and invasiveness. Several studies have also found a close relationship between cancer stem cells (CSCs) and biological features of metastasis, and the CSC markers ALDH1, ABCG2 and c-Met are expressed at high levels in metastatic PDAC cells. Emerging evidence reveals that exosomes are involved in the modulation of the tumor microenvironment and can initiate PDAC pre-metastatic niche formation in the liver and lungs. In this review, we provide an overview of the role of all these pivotal factors in the metastatic behavior of PDAC, and discuss their potential exploitation in the clinic to improve current therapeutics and identify new drug targets.
Collapse
Affiliation(s)
- E Giovannetti
- Lab Medical Oncology, Dept. Medical Oncology, VU University Medical Center (VUmc), Amsterdam, The Netherlands; Cancer Pharmacology Lab, AIRC Start Up Unit, University of Pisa, Pisa, Italy
| | - C L van der Borden
- Lab Medical Oncology, Dept. Medical Oncology, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | - A E Frampton
- HPB Surgical Unit, Dept. of Surgery & Cancer, Imperial College, Hammersmith Hospital Campus, London, UK
| | - A Ali
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, KP, Pakistan; Institute of Cancer Sciences, University of Glasgow, UK
| | - O Firuzi
- Lab Medical Oncology, Dept. Medical Oncology, VU University Medical Center (VUmc), Amsterdam, The Netherlands; Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - G J Peters
- Lab Medical Oncology, Dept. Medical Oncology, VU University Medical Center (VUmc), Amsterdam, The Netherlands.
| |
Collapse
|
14
|
CD44 Variant 6 as a Predictive Biomarker for Distant Metastasis in Patients With Epithelial Ovarian Cancer. Obstet Gynecol 2017; 127:1003-1011. [PMID: 27159753 DOI: 10.1097/aog.0000000000001420] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To investigate the role of cancer stem cell marker CD44 variant 6 in the development of distant metastasis in patients with epithelial ovarian cancer. METHODS A retrospective cohort study was performed in 186 patients who underwent surgery for ovarian cancer from 2005 to 2013 at the Kumamoto University Hospital. The association between the expression of CD44 variant 6 and distant metastasis was evaluated based on a large-scale immunohistochemical analysis. Primary ovarian tumors that contained at least 10% CD44 variant 6-positive cancer cells were categorized as CD44v6-high (n=53), and the tumors that contained less than 10% CD44 variant 6-positive cells were categorized as CD44v6-low (n=133). Distant metastasis-free survival was compared between the CD44v6-high and -low groups. Multivariate analysis was performed to estimate the influence of various clinicopathologic factors on the development of distant metastasis. RESULTS At the time of ovarian cancer diagnosis, distant metastasis occurred in 13 of 53 patients (24.5%) in the CD44v6-high group and 17 of 133 patients (12.8%) in the CD44v6-low group (P=.049). The median metastasis-free survival after stage I-III ovarian cancer diagnosis was 19.5 months (range 11-55 months) in the CD44v6-high group (n=40) and 39.5 months (range 22-57 months) in the CD44v6-low group (n=116) (P=.071). Multivariate analysis demonstrated that CD44 variant 6 expression was an independent risk factor for distant metastatic recurrence (hazard ratio 4.09, 95% confidence interval 1.29-12.98; P=.017). CONCLUSION CD44 variant 6 represents an important predictor of distant metastasis and CD44 variant 6-positive ovarian cancer cells play a crucial role in the formation of distant metastases in patients with ovarian cancer.
Collapse
|
15
|
Chen Z, Zhang L, Zhu Q, Wang X, Wu J, Wang X. Clinical value of octamer-binding transcription factor 4 as a prognostic marker in patients with digestive system cancers: A systematic review and meta-analysis. J Gastroenterol Hepatol 2017; 32:567-576. [PMID: 28320060 DOI: 10.1111/jgh.13624] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 09/10/2016] [Accepted: 09/27/2016] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND AIM The role of octamer-binding transcription factor 4 (Oct4) has been implicated in the clinical prognosis of various kinds of digestive system cancers, but the results remain controversial. The purpose of this meta-analysis is to assess the potential role of Oct4 as a prognostic marker in digestive system tumors. METHODS Relevant articles were retrieved from Pubmed, Web of Science, and Cochrane Library up to July 2016. The software Stata 12.0 was used to analyze the outcomes, including overall survival (OS), disease-free survival, recurrence-free survival, and clinicopathological characteristics. RESULTS A total of 13 eligible studies with 1538 patients were included. Elevated Oct4 expression was significantly associated with poor OS (pooled hazard ratio [HR] = 2.183, 95% confidence interval [CI]: 1.824-2.612), disease-free survival (pooled HR = 1.973, 95% CI: 1.538-2.532), and recurrence-free survival (pooled HR = 2.209, 95% CI: 1.461-3.338) of digestive system malignancies. Subgroup analyses showed that cancer type, sample size, study quality, and laboratory detection method did not alter the significant prognostic value of Oct4. Additionally, Oct4 expression was found to be an independent predictive factor for OS (HR = 2.068, 95% CI: 1.633-2.619). No significant association was found between Oct4 and clinicopathological features of digestive system malignancies. CONCLUSION This study provided evidence of Oct4 and/or its closely related homolog protein as a predictive factor for patients with digestive system cancers. More large-scale clinical studies on the prognostic value of Oct4 are warranted.
Collapse
Affiliation(s)
- Zhiqiang Chen
- Key Laboratory on Living Donor Liver Transplantation of Ministry of Health, Department of Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Long Zhang
- Key Laboratory on Living Donor Liver Transplantation of Ministry of Health, Department of Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qin Zhu
- Key Laboratory on Living Donor Liver Transplantation of Ministry of Health, Department of Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaowei Wang
- Key Laboratory on Living Donor Liver Transplantation of Ministry of Health, Department of Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jindao Wu
- Key Laboratory on Living Donor Liver Transplantation of Ministry of Health, Department of Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xuehao Wang
- Key Laboratory on Living Donor Liver Transplantation of Ministry of Health, Department of Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
16
|
Banerjee J, Papu John AM, Al-Wadei MH, Schuller HM. Prevention of pancreatic cancer in a hamster model by cAMP decrease. Oncotarget 2016; 7:44430-44441. [PMID: 27281617 PMCID: PMC5190108 DOI: 10.18632/oncotarget.9790] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 05/22/2016] [Indexed: 02/06/2023] Open
Abstract
Smoking and alcoholism are risk factors for the development of pancreatitis-associated pancreatic ductal adenocarcinoma (PDAC). We have previously shown that these cancers overexpressed stress neurotransmitters and cyclic adenosine monophosphate (cAMP) while the inhibitory neurotransmitter γ-aminobutyric acid (GABA) was suppressed. Using a hamster model, the current study has tested the hypothesis that cAMP decrease by GABA supplementation in the drinking water prevents the development of pancreatitis-associated PDAC. Our data reveal strong preventive effects of GABA supplementation on the development of PDAC and pancreatic intraductal neoplasia (PanIN). ELISA assays and immunohistochemistry revealed significant decreases in the levels of cAMP and interleukin 6 accompanied by reductions in the expression of several cancer stem cell markers and phosphorylated signaling proteins, which stimulate cell proliferation, and migration in pancreatic exocrine cells of GABA treated animals. We conclude that cAMP decrease by GABA supplementation inhibits multiple cancer stimulating pathways in cancer stem cells, differentiated cancer cells and the immune system, identifying this approach as promising novel tool for the prevention of PDAC in individuals with a history of smoking and alcoholism.
Collapse
Affiliation(s)
- Jheelam Banerjee
- Experimental Oncology Laboratory, Department of Biomedical & Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA
| | - Arokya M.S. Papu John
- Experimental Oncology Laboratory, Department of Biomedical & Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA
| | - Mohammed H. Al-Wadei
- Experimental Oncology Laboratory, Department of Biomedical & Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA
| | - Hildegard M. Schuller
- Experimental Oncology Laboratory, Department of Biomedical & Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA
| |
Collapse
|
17
|
Pei X, Zhu J, Yang R, Tan Z, An M, Shi J, Lubman DM. CD90 and CD24 Co-Expression Is Associated with Pancreatic Intraepithelial Neoplasias. PLoS One 2016; 11:e0158021. [PMID: 27332878 PMCID: PMC4917090 DOI: 10.1371/journal.pone.0158021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/08/2016] [Indexed: 12/28/2022] Open
Abstract
Thy-1 (CD90) has been shown to be a potential marker for several different types of cancer. However, reports on CD90 expression in pancreatic intraepithelial neoplasia (PanIN) lesions are still limited where PanINs are the most important precursor lesion of pancreatic ductal adenocarcinoma (PDAC). Herein, we investigate candidate markers for PanIN lesions by examining the distribution and trend of CD90 and CD24 expression as well as their co-expression in various stages of PanINs. Thirty cases of PanINs, which were confirmed histopathologically and clinically, were used to evaluate protein expression of CD90 and CD24 by immunofluoresence double staining. CD90 was found to be mainly expressed in stroma around lesion ducts while not observed in acini and islets in PanINs. CD90 also showed increased expression in PanIN III compared to PanIN III. CD24 was mainly present in the cytoplasm and membrane of pancreatic ductal epithelia, especially in the apical epithelium of the duct. CD24 had higher expression in PanIN III compared with PanIN IIIIII or PanIN III. CD90 was expressed around CD24 sites, but there was little overlap between cells that expressed each of these proteins. A correlation analysis showed that these two proteins have a moderate relationship with PanIN stages respectively. These results suggest that co-expression of CD90 and CD24 may have an important role in the development and progression of PanINs, which is also conducive to early detection and treatment of PDAC.
Collapse
Affiliation(s)
- Xiucong Pei
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan, 48109, United States of America
- Department of Toxicology, School of Public Health, Shenyang Medical College, Liaoning, 110034, China
| | - Jianhui Zhu
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan, 48109, United States of America
| | - Rui Yang
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan, 48109, United States of America
| | - Zhijing Tan
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan, 48109, United States of America
| | - Mingrui An
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan, 48109, United States of America
| | - Jiaqi Shi
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, 48109, United States of America
| | - David M. Lubman
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan, 48109, United States of America
| |
Collapse
|
18
|
Tyagi N, Marimuthu S, Bhardwaj A, Deshmukh SK, Srivastava SK, Singh AP, McClellan S, Carter JE, Singh S. p-21 activated kinase 4 (PAK4) maintains stem cell-like phenotypes in pancreatic cancer cells through activation of STAT3 signaling. Cancer Lett 2016; 370:260-267. [PMID: 26546043 PMCID: PMC4684758 DOI: 10.1016/j.canlet.2015.10.028] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 10/26/2015] [Accepted: 10/27/2015] [Indexed: 02/06/2023]
Abstract
Pancreatic cancer (PC) remains a highly lethal malignancy due to its unusual chemoresistance and high aggressiveness. A subpopulation of pancreatic tumor cells, known as cancer stem cells (CSCs), is considered responsible not only for tumor-maintenance, but also for its widespread metastasis and therapeutic failure. Here we investigated the role of p-21 activated kinase 4 (PAK4) in driving PC stemness properties. Our data demonstrate that triple-positive (CD24(+)/CD44(+)/EpCAM(+)) subpopulation of pancreatic CSCs exhibits greater level of PAK4 as compared to triple-negative (CD24(-)/CD44(-)/EpCAM(-)) cells. Moreover, PAK4 silencing in PC cells leads to diminished fraction of CD24, CD44, and EpCAM positive cells. Furthermore, we show that PAK4-silenced PC cells exhibit decreased sphere-forming ability and increased chemosensitivity to gemcitabine toxicity. PAK4 expression is also associated with enhanced levels of stemness-associated transcription factors (Oct4/Nanog/Sox2 and KLF4). Furthermore, our data show decreased nuclear accumulation and transcriptional activity of STAT3 in PAK4-silenced PC cells and restitution of its activity leads to restoration of stem cell phenotypes. Together, our findings deliver first experimental evidence for the involvement of PAK4 in PC stemness and support its clinical utility as a novel therapeutic target in PC.
Collapse
Affiliation(s)
- Nikhil Tyagi
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama 36604, USA
| | - Saravanakumar Marimuthu
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama 36604, USA
| | - Arun Bhardwaj
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama 36604, USA
| | - Sachin K Deshmukh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama 36604, USA
| | - Sanjeev K Srivastava
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama 36604, USA
| | - Ajay P Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama 36604, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama 36688, USA
| | - Steven McClellan
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama 36604, USA
| | - James E Carter
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama 36688, USA
| | - Seema Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama 36604, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama 36688, USA.
| |
Collapse
|
19
|
Salaria S, Means A, Revetta F, Idrees K, Liu E, Shi C. Expression of CD24, a Stem Cell Marker, in Pancreatic and Small Intestinal Neuroendocrine Tumors. Am J Clin Pathol 2015; 144:642-8. [PMID: 26386086 DOI: 10.1309/ajcpmzy5p9twnjjv] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES CD24 has been considered a normal and cancer stem cell marker. Potential intestinal stem cells weakly express CD24. In the pancreas, CD24 is a possible cancer stem cell marker for ductal adenocarcinoma. METHODS Expression of CD24 in intestinal and pancreatic neuroendocrine tumors (NETs) was examined. Immunohistochemistry was performed on benign duodenum, ileum mucosa, and pancreas, as well as primary duodenal, primary and metastatic ileal, and pancreatic NETs. RESULTS Scattered CD24-positive cells were noted in the duodenal and ileal crypts, most of which showed a strong subnuclear labeling pattern. Similar expression was observed in 41 (95%) of 43 primary ileal NETs but in only four (15%) of 26 duodenal NETs (P < .01). In addition, metastatic ileal NETs retained CD24 expression. Pancreatic islets did not express CD24, and only rare cells had subnuclear labeling of CD24 in the pancreatic ducts. Unlike ileal NETs, only five (5%) of 92 pancreatic NETs expressed CD24 in the subnuclear compartment (P < .01). All five NETs showed a unique morphology with prominent stromal fibrosis. CONCLUSIONS CD24 expression was frequent in primary and metastatic midgut NETs but rare in pancreatic and duodenal NETs. Expression of CD24 in ileal NETs may have future diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Safia Salaria
- Departments of Pathology, Microbiology, and Immunology
| | - Anna Means
- Surgery, Vanderbilt University Hospital, Nashville, TN
| | - Frank Revetta
- Departments of Pathology, Microbiology, and Immunology
| | - Kamran Idrees
- Surgical Oncology, Vanderbilt University Hospital, Nashville, TN
| | - Eric Liu
- Surgical Oncology, Vanderbilt University Hospital, Nashville, TN
| | - Chanjuan Shi
- Departments of Pathology, Microbiology, and Immunology
| |
Collapse
|
20
|
WANG WENJIE, WU MENGYAO, SHEN MENG, ZHI QIAOMING, LIU ZEYI, GONG FEIRAN, TAO MIN, LI WEI. Cantharidin and norcantharidin impair stemness of pancreatic cancer cells by repressing the β-catenin pathway and strengthen the cytotoxicity of gemcitabine and erlotinib. Int J Oncol 2015; 47:1912-22. [DOI: 10.3892/ijo.2015.3156] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/20/2015] [Indexed: 11/06/2022] Open
|
21
|
Tanaka T, Terai Y, Kogata Y, Ashihara K, Maeda K, Fujiwara S, Yoo S, Tanaka Y, Tsunetoh S, Sasaki H, Kanemura M, Tanabe A, Ohmichi M. CD24 expression as a marker for predicting clinical outcome and invasive activity in uterine cervical cancer. Oncol Rep 2015; 34:2282-8. [PMID: 26351781 PMCID: PMC4583540 DOI: 10.3892/or.2015.4257] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 07/08/2015] [Indexed: 01/06/2023] Open
Abstract
CD24, a small heavily glycosylated mucin-like glycosylphosphatidylinositol-anchored cell surface protein, plays an important role in the carcinogenesis of various human malignancies. However, its function in cervical cancer remains unclear. The aim of the present study was to evaluate the expression of CD24 clinicopathologically and to analyze its functional behavior biologically in cervical cancer. A total of 117 uterine cervical cancer tumors were immunohistochemically analyzed using a CD24 monoclonal antibody on paraffin blocks. We also examined whether CD24 enhanced the invasive activity or the Akt, ERK, NF-κB and MMP activity in a uterine cervical cancer cell line (CaSki) by a western blot analysis. The patients with enhanced CD24 expression had a higher rate of advanced clinical stage (50 vs. 16.5%, p<0.01), lymph node metastasis (34.6 vs. 14.3%) and lymphovascular involvement (65.4 vs. 20.4%, p=0.01), and a poor overall and disease-free survival (5-year survival rate: 62 vs. 86%, p=0.03). CD24 overexpression in CaSki cells resulted in activation of Cell Signaling proteins, including Akt, ERK, NF-κB and MMP-9. An invasion assay showed that CD24 overexpression in CaSki cells led to increased invasion ability. The CD24 overexpression also increased mRNA expression of Slug but not Snail. Moreover, the CD24 overexpression also decreased expression of E-cadherin and increased N-cadherin protein levels. Increased expression of CD24 may be associated with tumor progression and prognosis in patients with uterine cervical cancer. CD24 expression may therefore be used not only as a prognostic marker in uterine cervical cancer, but also as a target for the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Tomohito Tanaka
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Yoshito Terai
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Yuhei Kogata
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Keisuke Ashihara
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Kazuya Maeda
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Satoe Fujiwara
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Saha Yoo
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Yoshimichi Tanaka
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Satoshi Tsunetoh
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Hiroshi Sasaki
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Masanori Kanemura
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Akiko Tanabe
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Masahide Ohmichi
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| |
Collapse
|
22
|
Okada M, Shibuya K, Sato A, Seino S, Suzuki S, Seino M, Kitanaka C. Targeting the K-Ras--JNK axis eliminates cancer stem-like cells and prevents pancreatic tumor formation. Oncotarget 2015; 5:5100-12. [PMID: 24947996 PMCID: PMC4148125 DOI: 10.18632/oncotarget.2087] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cancer cells with self-renewal and tumor-initiating capacity, either quiescent (cancer stem cells, CSCs) or proliferating (cancer stem-like cells, CSLCs), are now deemed responsible for the pervasive therapy resistance of pancreatic cancer, one of the deadliest human cancers characterized by high prevalence of K-Ras mutation. However, to date, much remains unknown how pancreatic CSCs/CSLCs are regulated. Here we show that the K-Ras – JNK axis plays a pivotal role in the maintenance of pancreatic CSCs/CSLCs. In vitro inhibition of JNK, either pharmacological or genetic, caused loss of the self-renewal and tumor-initiating capacity of pancreatic CSLCs. Importantly, JNK inhibition in vivo via systemic JNK inhibitor administration, which had no discernible effect on the general health status of mice, efficiently depleted the CSC/CSLC population within pre-established pancreatic tumor xenografts. Furthermore, knockdown of K-Ras in pancreatic CSLCs with K-Ras mutation led to downregulation of the JNK pathway as well as in loss of self-renewal and tumor-initiating capacity. Together, our findings suggest that pancreatic CSCs/CSLCs are dependent on K-Ras activation of JNK and also suggest that the K-Ras – JNK axis could be a potential target in CSC/CSLC-directed therapies against pancreatic cancer.
Collapse
Affiliation(s)
- Masashi Okada
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan
| | | | | | | | | | | | - Chifumi Kitanaka
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan; Oncology Research Center, Research Institute for Advanced Molecular Epidemiology, Yamagata University, Yamagata, Japan; Global COE program for Medical Sciences, Japan Society for Promotion of Science, Tokyo, Japan; Research Institute for Promotion of Medical Sciences, Yamagata University School of Medicine, Yamagata, Japan
| |
Collapse
|
23
|
Dalla Pozza E, Dando I, Biondani G, Brandi J, Costanzo C, Zoratti E, Fassan M, Boschi F, Melisi D, Cecconi D, Scupoli MT, Scarpa A, Palmieri M. Pancreatic ductal adenocarcinoma cell lines display a plastic ability to bi‑directionally convert into cancer stem cells. Int J Oncol 2015; 46:1099-1108. [PMID: 25502497 DOI: 10.3892/ijo.2014.2796] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 11/18/2014] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is often diagnosed when metastatic events have occurred. Cancer stem cells (CSCs) play an important role in tumor initiation, metastasis, chemoresistance and relapse. A growing number of studies have suggested that CSCs exist in a dynamic equilibrium with more differentiated cancer cells via a bi‑directional regeneration that is dependent on the environmental stimuli. In this investigation, we obtain, by using a selective medium, PDAC CSCs from five out of nine PDAC cell lines, endowed with different tumorsphere‑forming ability. PDAC CSCs were generally more resistant to the action of five anticancer drugs than parental cell lines and were characterized by an increased expression of EpCAM and CD44v6, typical stem cell surface markers, and a decreased expression of E‑cadherin, the main marker of the epithelial state. PDAC CSCs were able to re‑differentiate into parental cells once cultured in parental growth condition, as demonstrated by re‑acquisition of the epithelial morphology, the decreased expression levels of EpCAM and CD44v6 and the increased sensitivity to anticancer drugs. Finally, PDAC CSCs injected into nude mice developed a larger subcutaneous tumor mass and showed a higher metastatic activity compared to parental cells. The present study demonstrates the ability to obtain CSCs from several PDAC cell lines and that these cells are differentially resistant to various anticancer agents. This variability renders them a model of great importance to deeply understand pancreatic adenocarcinoma biology, to discover new biomarkers and to screen new therapeutic compounds.
Collapse
Affiliation(s)
- Elisa Dalla Pozza
- Department of Life and Reproduction Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Ilaria Dando
- Department of Life and Reproduction Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Giulia Biondani
- Department of Life and Reproduction Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Jessica Brandi
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Chiara Costanzo
- Department of Life and Reproduction Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Elisa Zoratti
- Applied Research on Cancer Network (ARC‑NET) and Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Matteo Fassan
- Applied Research on Cancer Network (ARC‑NET) and Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Federico Boschi
- Department of Computer Science, University of Verona, Verona, Italy
| | - Davide Melisi
- Department of Medicine, Oncology Unit, University and Hospital Trust of Verona, Verona, Italy
| | - Daniela Cecconi
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Maria Teresa Scupoli
- Applied Research on Cancer Network (ARC‑NET) and Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Aldo Scarpa
- Applied Research on Cancer Network (ARC‑NET) and Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Marta Palmieri
- Department of Life and Reproduction Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| |
Collapse
|
24
|
Zhan HX, Xu JW, Wu D, Zhang TP, Hu SY. Pancreatic cancer stem cells: new insight into a stubborn disease. Cancer Lett 2015; 357:429-37. [PMID: 25499079 DOI: 10.1016/j.canlet.2014.12.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Revised: 11/30/2014] [Accepted: 12/02/2014] [Indexed: 02/07/2023]
Abstract
Resistance to conventional therapy and early distant metastasis contribute to the unsatisfactory prognosis of patients with pancreatic cancer. The concept of cancer stem cells (CSCs) brings new insights into cancer biology and therapy. Many studies have confirmed the important role of these stem cells in carcinogenesis and the development of hematopoietic and solid cancers. Recent studies have shown that CSCs regulate aggressive behavior, recurrence, and drug resistance in pancreatic cancer. Here, we review recent advances in pancreatic cancer stem cells (PCSCs) research. Particular attention is paid to the regulation mechanisms of pancreatic cancer stem cell functions, such as stemness-related signaling pathways, microRNAs, the epithelial-mesenchymal transition (EMT), and the tumor microenvironment, and the development of novel PCSCs targeted therapy. We seek to further understand PCSCs and explore potential therapeutic targets for pancreatic cancer.
Collapse
Affiliation(s)
- Han-xiang Zhan
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Jian-wei Xu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Dong Wu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Tai-ping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - San-yuan Hu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China.
| |
Collapse
|
25
|
Pan Y, Gao S, Hua YQ, Liu LM. Establishment of a Pancreatic Cancer Stem Cell Model Using the SW1990 Human Pancreatic Cancer Cell Line in Nude Mice. Asian Pac J Cancer Prev 2015; 16:437-42. [DOI: 10.7314/apjcp.2015.16.2.437] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
26
|
Bao B, Azmi AS, Ali S, Zaiem F, Sarkar FH. Metformin may function as anti-cancer agent via targeting cancer stem cells: the potential biological significance of tumor-associated miRNAs in breast and pancreatic cancers. ANNALS OF TRANSLATIONAL MEDICINE 2014; 2:59. [PMID: 25333034 DOI: 10.3978/j.issn.2305-5839.2014.06.05] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 05/27/2014] [Indexed: 12/13/2022]
Abstract
Metformin is one of the most used diabetic drugs for the management of type II diabetes mellitus (DM) in the world. Increased numbers of epidemiological and clinical studies have provided convincing evidence supporting the role of metformin in the development and progression of a variety of human tumors including breast and pancreatic cancer. Substantial pre-clinical evidence from in vitro and in vivo experimental studies strongly suggests that metformin has an anti-cancer activity mediated through the regulation of several cell signaling pathways including activation of AMP kinase (AMPK), and other direct and indirect mechanisms; however, the detailed mechanism(s) has not yet been fully understood. The concept of cancer stem cells (CSCs) has gained significant attention in recent years due its identification and defining its clinical implications in many different tumors including breast cancer and pancreatic cancer. In this review, we will discuss the protective role of metformin in the development of breast and pancreatic cancers. We will further discuss the role of metformin as an anti-cancer agent, which is in part mediated through targeting CSCs. Finally, we will discuss the potential role of metformin in the modulation of tumor-associated or CSC-associated microRNAs (miRNAs) as part of the novel mechanism of action of metformin in the development and progression of breast and pancreatic cancers.
Collapse
Affiliation(s)
- Bin Bao
- 1 Department of Pathology, 2 Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Asfar S Azmi
- 1 Department of Pathology, 2 Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Shadan Ali
- 1 Department of Pathology, 2 Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Feras Zaiem
- 1 Department of Pathology, 2 Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Fazlul H Sarkar
- 1 Department of Pathology, 2 Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
27
|
Single-cell RNA sequencing identifies extracellular matrix gene expression by pancreatic circulating tumor cells. Cell Rep 2014. [PMID: 25242334 DOI: 10.1016/j.celrep.2014.08.029.single-cell] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Circulating tumor cells (CTCs) are shed from primary tumors into the bloodstream, mediating the hematogenous spread of cancer to distant organs. To define their composition, we compared genome-wide expression profiles of CTCs with matched primary tumors in a mouse model of pancreatic cancer, isolating individual CTCs using epitope-independent microfluidic capture, followed by single-cell RNA sequencing. CTCs clustered separately from primary tumors and tumor-derived cell lines, showing low-proliferative signatures, enrichment for the stem-cell-associated gene Aldh1a2, biphenotypic expression of epithelial and mesenchymal markers, and expression of Igfbp5, a gene transcript enriched at the epithelial-stromal interface. Mouse as well as human pancreatic CTCs exhibit a very high expression of stromal-derived extracellular matrix (ECM) proteins, including SPARC, whose knockdown in cancer cells suppresses cell migration and invasiveness. The aberrant expression by CTCs of stromal ECM genes points to their contribution of microenvironmental signals for the spread of cancer to distant organs.
Collapse
|
28
|
Ting DT, Wittner BS, Ligorio M, Vincent Jordan N, Shah AM, Miyamoto DT, Aceto N, Bersani F, Brannigan BW, Xega K, Ciciliano JC, Zhu H, MacKenzie OC, Trautwein J, Arora KS, Shahid M, Ellis HL, Qu N, Bardeesy N, Rivera MN, Deshpande V, Ferrone CR, Kapur R, Ramaswamy S, Shioda T, Toner M, Maheswaran S, Haber DA. Single-cell RNA sequencing identifies extracellular matrix gene expression by pancreatic circulating tumor cells. Cell Rep 2014; 8:1905-1918. [PMID: 25242334 PMCID: PMC4230325 DOI: 10.1016/j.celrep.2014.08.029] [Citation(s) in RCA: 381] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 07/16/2014] [Accepted: 08/13/2014] [Indexed: 12/27/2022] Open
Abstract
Circulating tumor cells (CTCs) are shed from primary tumors into the bloodstream, mediating the hematogenous spread of cancer to distant organs. To define their composition, we compared genome-wide expression profiles of CTCs with matched primary tumors in a mouse model of pancreatic cancer, isolating individual CTCs using epitope-independent microfluidic capture, followed by single-cell RNA sequencing. CTCs clustered separately from primary tumors and tumor-derived cell lines, showing low-proliferative signatures, enrichment for the stem-cell-associated gene Aldh1a2, biphenotypic expression of epithelial and mesenchymal markers, and expression of Igfbp5, a gene transcript enriched at the epithelial-stromal interface. Mouse as well as human pancreatic CTCs exhibit a very high expression of stromal-derived extracellular matrix (ECM) proteins, including SPARC, whose knockdown in cancer cells suppresses cell migration and invasiveness. The aberrant expression by CTCs of stromal ECM genes points to their contribution of microenvironmental signals for the spread of cancer to distant organs.
Collapse
Affiliation(s)
- David T Ting
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Ben S Wittner
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Matteo Ligorio
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Surgery, Harvard Medical School, Boston, MA 02114, USA; Department of Health Sciences, University of Genoa, 16126 Genoa, Italy
| | - Nicole Vincent Jordan
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Ajay M Shah
- Center for Engineering in Medicine, Harvard Medical School, Boston, MA 02114, USA; Department of Surgery, Harvard Medical School, Boston, MA 02114, USA
| | - David T Miyamoto
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Radiation Oncology, Harvard Medical School, Boston, MA 02114, USA
| | - Nicola Aceto
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Francesca Bersani
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Brian W Brannigan
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Kristina Xega
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Jordan C Ciciliano
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Huili Zhu
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Olivia C MacKenzie
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Julie Trautwein
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Kshitij S Arora
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Surgery, Harvard Medical School, Boston, MA 02114, USA; Department of Pathology, Harvard Medical School, Boston, MA 02114, USA
| | - Mohammad Shahid
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Surgery, Harvard Medical School, Boston, MA 02114, USA; Department of Pathology, Harvard Medical School, Boston, MA 02114, USA
| | - Haley L Ellis
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Na Qu
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Nabeel Bardeesy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Miguel N Rivera
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Pathology, Harvard Medical School, Boston, MA 02114, USA
| | - Vikram Deshpande
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Pathology, Harvard Medical School, Boston, MA 02114, USA
| | - Cristina R Ferrone
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Surgery, Harvard Medical School, Boston, MA 02114, USA
| | - Ravi Kapur
- Center for Engineering in Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Sridhar Ramaswamy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Toshi Shioda
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Mehmet Toner
- Center for Engineering in Medicine, Harvard Medical School, Boston, MA 02114, USA; Department of Surgery, Harvard Medical School, Boston, MA 02114, USA
| | - Shyamala Maheswaran
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Surgery, Harvard Medical School, Boston, MA 02114, USA.
| | - Daniel A Haber
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
29
|
Fitzgerald TL, McCubrey JA. Pancreatic cancer stem cells: association with cell surface markers, prognosis, resistance, metastasis and treatment. Adv Biol Regul 2014; 56:45-50. [PMID: 24925031 DOI: 10.1016/j.jbior.2014.05.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 05/07/2014] [Accepted: 05/12/2014] [Indexed: 02/07/2023]
Abstract
In this review, we will discuss the recent advances in understanding the pancreatic cancer stem cells. Identification and characterization of pancreatic cancer stem cells may aid our ability to improve diagnosis and treatment of pancreatic cancer. Novel approaches are necessary for the earlier diagnosis of pancreatic cancer as well as improved treatment to prevent distal metastasis. Key markers for the identification of pancreatic cancer stem cells include CD133, ALDH, side population cells and the triplet combination CD44+ CD24+ESA+. The roles of these proteins as markers for stemness in pancreatic cancer as well as recent studies with the c-Met proto-oncogene will be discussed. The ability of these markers to predict survival of pancreatic cancer patients is being examined clinically. Stemness and resistance to chemotherapy and radiotherapy may be linked. Expression of some of these markers may be associated with distant metastasis. Treatment of pancreatic cancer patients by targeting the pancreatic cancer stem cells holds promise.
Collapse
Affiliation(s)
- Timothy L Fitzgerald
- Department of Surgery, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
30
|
Tanase CP, Neagu AI, Necula LG, Mambet C, Enciu AM, Calenic B, Cruceru ML, Albulescu R. Cancer stem cells: involvement in pancreatic cancer pathogenesis and perspectives on cancer therapeutics. World J Gastroenterol 2014; 20:10790-10801. [PMID: 25152582 PMCID: PMC4138459 DOI: 10.3748/wjg.v20.i31.10790] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 02/07/2014] [Accepted: 04/05/2014] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the most aggressive and lethal malignancies. Despite remarkable progress in understanding pancreatic carcinogenesis at the molecular level, as well as progress in new therapeutic approaches, pancreatic cancer remains a disease with a dismal prognosis. Among the mechanisms responsible for drug resistance, the most relevant are changes in individual genes or signaling pathways and the presence of highly resistant cancer stem cells (CSCs). In pancreatic cancer, CSCs represent 0.2%-0.8% of pancreatic cancer cells and are considered to be responsible for tumor growth, invasion, metastasis and recurrence. CSCs have been extensively studied as of late to identify specific surface markers to ensure reliable sorting and for signaling pathways identified to play a pivotal role in CSC self-renewal. Involvement of CSCs in pancreatic cancer pathogenesis has also highlighted these cells as the preferential targets for therapy. The present review is an update of the results in two main fields of research in pancreatic cancer, pathogenesis and therapy, focused on the narrow perspective of CSCs.
Collapse
|
31
|
The impact of Aldehyde dehydrogenase 1 expression on prognosis for metastatic colon cancer. J Surg Res 2014; 192:82-9. [PMID: 24953984 DOI: 10.1016/j.jss.2014.05.054] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 04/20/2014] [Accepted: 05/16/2014] [Indexed: 12/30/2022]
Abstract
BACKGROUND Cancer stem cells may be associated with tumor progression and prognosis for colon cancer. We hypothesized that expression of Aldehyde dehydrogenase 1 (ALDH1) would increase with tumor progression and be associated with survival. METHODS Tissue was obtained from resection specimens for isolation of cancer stem cells. In addition, paraffin blocks from resected colon cancers with normal colon, primary tumor, and lymph node and liver metastasis from 2000 to 2010 were identified and stained with ALDH1. RESULTS In in vitro models (adherent and tumor spheres) ALHD1+ cells grew more efficiently than ALDH1- cells. ALDH1 expression was highest in peritumoral crypt cells (0.137 μm(2), 95% confidence interval [CI] 0.125-0.356) and normal crypts (median 0.091 μm(2), 95% CI 0.064-0.299) followed by lymph node metastasis (median 0.025 μm(2), 95% CI 0-0.131) and the primary cancers (median 0.014 μm(2), 95% CI 0.0123-0.154). Samples were divided into high and low ALDH1 expression. Survival was associated with expression in the primary tumor (9 versus 23 mo, P = 0.0016) expression but not peritumoral tissue (21 versus 20.5 mo, P = 0.32), normal colon (19 versus 27 mo, P = 0.289), or lymph node metastasis (23 versus 21 mo, P = 0.69). On univariate analysis, ALDH1 expression and grade were associated with survival but ages, number of lymph node metastasis, race, or grade were not associated. On multivariate analysis, only ALDH1 status continued to be associated with survival, odds ratio 4.4, and P = 0.011. CONCLUSIONS ALDH1 is indicative of stemness and is a biomarker marker in colon cancer. Expression did not increase with progression from normal colon to primary tumors and metastasis.
Collapse
|
32
|
Polvani S, Tarocchi M, Tempesti S, Mello T, Ceni E, Buccoliero F, D'Amico M, Boddi V, Farsi M, Nesi S, Nesi G, Milani S, Galli A. COUP-TFII in pancreatic adenocarcinoma: clinical implication for patient survival and tumor progression. Int J Cancer 2014; 134:1648-58. [PMID: 24122412 DOI: 10.1002/ijc.28502] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 08/10/2013] [Accepted: 09/06/2013] [Indexed: 12/16/2023]
Abstract
Despite the accumulating knowledge of alterations in pancreatic cancer molecular pathways, no substantial improvements in the clinical prognosis have been made and this malignancy continues to be a leading cause of cancer death in the Western World. The orphan nuclear receptor COUP-TFII is a regulator of a wide range of biological processes and it may exert a pro-oncogenic role in cancer cells; interestingly, indirect evidences suggest that the receptor could be involved in pancreatic cancer. The aim of this study was to evaluate the expression of COUP-TFII in human pancreatic tumors and to unveil its role in the regulation of pancreatic tumor growth. We evaluated COUP-TFII expression by immunohistochemistry on primary samples. We analyzed the effect of the nuclear receptor silencing in human pancreatic cancer cells by means of shRNA expressing cell lines. We finally confirmed the in vitro results by in vivo experiments on nude mice. COUP-TFII is expressed in 69% of tested primary samples and correlates with the N1 and M1 status and clinical stage; Kaplan-Meier and Cox regression analysis show that it may be an independent prognostic factor of worst outcome. In vitro silencing of COUP-TFII reduces the cell growth and invasiveness and it strongly inhibits angiogenesis, an effect mediated by the regulation of VEGF-C. In nude mice, COUP-TFII silencing reduces tumor growth by 40%. Our results suggest that COUP-TFII might be an important regulator of the behavior of pancreatic adenocarcinoma, thus representing a possible new target for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Simone Polvani
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Firenze, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Tumor-induced STAT3 activation in monocytic myeloid-derived suppressor cells enhances stemness and mesenchymal properties in human pancreatic cancer. Cancer Immunol Immunother 2014; 63:513-28. [PMID: 24652403 PMCID: PMC3994288 DOI: 10.1007/s00262-014-1527-x] [Citation(s) in RCA: 161] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Accepted: 02/22/2014] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer (PC) mobilizes myeloid cells from the bone marrow to the tumor where they promote tumor growth and proliferation. Cancer stem cells (CSCs) are a population of tumor cells that are responsible for tumor initiation. Aldehyde dehydrogenase-1 activity in PC identifies CSCs, and its activity has been correlated with poor overall prognosis in human PC. Myeloid cells have been shown to impact tumor stemness, but the impact of immunosuppressive tumor-infiltrating granulocytic and monocytic myeloid-derived suppressor cells (Mo-MDSC) on ALDH1Bright CSCs and epithelial to mesenchymal transition is not well understood. In this study, we demonstrate that Mo-MDSC (CD11b+/Gr1+/Ly6G−/Ly6Chi) significantly increase the frequency of ALDH1Bright CSCs in a mouse model of PC. Additionally, there was significant upregulation of genes associated with epithelial to mesenchymal transition. We also found that human PC converts CD14+ peripheral blood monocytes into Mo-MDSC (CD14+/HLA-DRlow/−) in vitro, and this transformation is dependent on the activation of the STAT3 pathway. In turn, these Mo-MDSC increase the frequency of ALDH1Bright CSCs and promote mesenchymal features of tumor cells. Finally, blockade of STAT3 activation reversed the increase in ALDH1Bright CSCs. These data suggest that the PC tumor microenvironment transforms monocytes to Mo-MDSC by STAT3 activation, and these cells increase the frequency of ALDH1Bright CSCs. Therefore, targeting STAT3 activation may be an effective therapeutic strategy in targeting CSCs in PC.
Collapse
|
34
|
Ji H, Greening DW, Barnes TW, Lim JW, Tauro BJ, Rai A, Xu R, Adda C, Mathivanan S, Zhao W, Xue Y, Xu T, Zhu HJ, Simpson RJ. Proteome profiling of exosomes derived from human primary and metastatic colorectal cancer cells reveal differential expression of key metastatic factors and signal transduction components. Proteomics 2013; 13:1672-86. [PMID: 23585443 DOI: 10.1002/pmic.201200562] [Citation(s) in RCA: 274] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 03/08/2013] [Accepted: 03/26/2013] [Indexed: 12/14/2022]
Abstract
Exosomes are small extracellular 40-100 nm diameter membrane vesicles of late endosomal origin that can mediate intercellular transfer of RNAs and proteins to assist premetastatic niche formation. Using primary (SW480) and metastatic (SW620) human isogenic colorectal cancer cell lines we compared exosome protein profiles to yield valuable insights into metastatic factors and signaling molecules fundamental to tumor progression. Exosomes purified using OptiPrep™ density gradient fractionation were 40-100 nm in diameter, were of a buoyant density ~1.09 g/mL, and displayed stereotypic exosomal markers TSG101, Alix, and CD63. A major finding was the selective enrichment of metastatic factors (MET, S100A8, S100A9, TNC), signal transduction molecules (EFNB2, JAG1, SRC, TNIK), and lipid raft and lipid raft-associated components (CAV1, FLOT1, FLOT2, PROM1) in exosomes derived from metastatic SW620 cells. Additionally, using cryo-electron microscopy, ultrastructural components in exosomes were identified. A key finding of this study was the detection and colocalization of protein complexes EPCAM-CLDN7 and TNIK-RAP2A in colorectal cancer cell exosomes. The selective enrichment of metastatic factors and signaling pathway components in metastatic colon cancer cell-derived exosomes contributes to our understanding of the cross-talk between tumor and stromal cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Hong Ji
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Bao B, Ahmad A, Azmi AS, Ali S, Sarkar FH. Overview of cancer stem cells (CSCs) and mechanisms of their regulation: implications for cancer therapy. ACTA ACUST UNITED AC 2013; Chapter 14:Unit 14.25. [PMID: 23744710 DOI: 10.1002/0471141755.ph1425s61] [Citation(s) in RCA: 182] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The identification of small subpopulations of cancer stem cells (CSCs) from blood mononuclear cells in human acute myeloid leukemia (AML) in 1997 was a landmark observation that recognized the potential role of CSCs in tumor aggressiveness. Two critical properties contribute to the functional role of CSCs in the establishment and recurrence of cancerous tumors: their capacity for self-renewal and their potential to differentiate into unlimited heterogeneous populations of cancer cells. These findings suggest that CSCs may represent novel therapeutic targets for the treatment and/or prevention of tumor progression, since they appear to be involved in cell migration, invasion, metastasis, and treatment resistance-all of which lead to poor clinical outcomes. The identification of CSC-specific markers, the isolation and characterization of CSCs from malignant tissues, and targeting strategies for the destruction of CSCs provide a novel opportunity for cancer research. This overview describes the potential implications of several common CSC markers in the identification of CSC subpopulations that are restricted to common malignant diseases, e.g., leukemia, and breast, prostate, pancreatic, and lung cancers. The role of microRNAs (miRNAs) in the regulation of CSC function is also discussed, as are several methods commonly used in CSC research. The potential role of the antidiabetic drug metformin- which has been shown to have effects on CSCs, and is known to function as an antitumor agent-is discussed as an example of this new class of chemotherapeutics.
Collapse
Affiliation(s)
- Bin Bao
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | | | | | | | | |
Collapse
|
36
|
Abourbih S, Sircar K, Tanguay S, Kassouf W, Aprikian A, Mansure J, Brimo F. Aldehyde dehydrogenase 1 expression in primary and metastatic renal cell carcinoma: an immunohistochemistry study. World J Surg Oncol 2013; 11:298. [PMID: 24266898 PMCID: PMC3842840 DOI: 10.1186/1477-7819-11-298] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 11/09/2013] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND ALDH1 has been shown to be a cancer stem cell marker, and its expression correlates with prognosis in a number of malignancies. We aimed to evaluate the expression of ALDH1 in a cohort of primary and metastatic RCC specimens, and to correlate expression with pathological outcomes such as tumor stage and grade, and clinical outcomes such as progression free survival. METHODS Three tissue microarrays were constructed from 244 RCC specimens, taken from 1985 to 2006. Samples were stained using an ALDH1 monoclonal antibody and expression was quantified by degree of staining. Membrane and cytoplasm staining were considered separately. A retrospective chart review enabled correlation with clinical outcomes. RESULTS ALDH1 expression did not vary significantly based on tumor stage (P = 0.6274) or grade (P = 0.1666). ALDH1 showed significantly more membranous expression in clear cell RCC versus other subtypes (P < 0.0001), as well as in the primary setting compared to metastases (P = 0.0216). In terms of progression free survival, no significant differences were seen based on ALDH1 expression levels. In a subanalysis of clear cell tumors, ALDH1 membranous expression was decreased in tumors of higher stage (P = 0.0233). CONCLUSIONS ALDH1 may be useful in characterizing RCC tumors as clear cell subtype. However, unlike in other malignancies, ALDH1 may not be useful in prognosticating renal cancers. The clinical significance of decreased ALDH1 expression in the high stage and metastatic setting remains to be determined in further investigations.
Collapse
Affiliation(s)
- Samuel Abourbih
- Department of Urology, McGill University Health Center, Montreal, QC, Canada
| | - Kanishka Sircar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Simon Tanguay
- Department of Urology, McGill University Health Center, Montreal, QC, Canada
| | - Wassim Kassouf
- Department of Urology, McGill University Health Center, Montreal, QC, Canada
| | - Armen Aprikian
- Department of Urology, McGill University Health Center, Montreal, QC, Canada
| | - Jose Mansure
- Department of Urology, McGill University Health Center, Montreal, QC, Canada
| | - Fadi Brimo
- Department of Pathology, McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
37
|
Castellanos JA, Merchant NB, Nagathihalli NS. Emerging targets in pancreatic cancer: epithelial-mesenchymal transition and cancer stem cells. Onco Targets Ther 2013; 6:1261-7. [PMID: 24049451 PMCID: PMC3775701 DOI: 10.2147/ott.s34670] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Pancreatic ductal adenocarcinoma is one of the most aggressive solid malignancies and is characterized by poor response to current therapy and a dismal survival rate. Recent insights regarding the role of cancer stem cells (CSCs) and epithelial–mesenchymal transition (EMT) in tumorigenesis have brought further understanding to the field and have highlighted new therapeutic targets. CSCs are a distinct subset of cancer cells, with the ability to differentiate into other cell types and self-renew in order to fuel the maintenance of tumor amplification. Transition of a cancer cell from an EMT leads to increased migratory and invasive properties, and thus facilitates initiation of metastasis. EMT is regulated by a complex network of factors that includes cytokines, growth factors, aberrant signaling pathways, transcription factors, and the tumor microenvironment. There is emerging evidence that the EMT process may give rise to CSCs, or at least cells with stem cell-like properties. We review the key pathways involved in both of these processes, the biomarkers used to identify CSCs, and new therapeutic approaches targeting CSCs and EMT in pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Jason A Castellanos
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | |
Collapse
|
38
|
Concomitant targeting of multiple key transcription factors effectively disrupts cancer stem cells enriched in side population of human pancreatic cancer cells. PLoS One 2013; 8:e73942. [PMID: 24040121 PMCID: PMC3770686 DOI: 10.1371/journal.pone.0073942] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 07/24/2013] [Indexed: 12/17/2022] Open
Abstract
Background A major challenge in the treatment of pancreatic ductal adenocarcinoma is the failure of chemotherapy, which is likely due to the presence of the cancer stem cells (CSCs). Objective To identify side population (SP) cells and characterize s-like properties in human pancreatic cancer cell lines (h-PCCLs) and to exploit the efficacy of concomitant targeting of multiple key transcription factors governing the stemness of pancreatic CSCs in suppressing CSC-like phenotypes. Methods Flow cytometry and Hoechst 33342 DNA-binding dye efflux assay were used to sort SP and non-SP (NSP) cells from three h-PCCLs: PANC-1, SW1990, and BxPc-3. The self-renewal ability, invasiveness, migration and drug resistance of SP cells were evaluated. Expression of CSC marker genes was analyzed. Tumorigenicity was assessed using a xenograft model in nude mice. Effects of a complex decoy oligonucleotide (cdODN-SCO) designed to simultaneously targeting Sox2, Oct4 and c-Myc were assessed. Results CSCs were enriched in the side proportion (SP) cells contained in the h-PCCLs and they possessed aggressive growth, invasion, migration and drug-resistance properties, compared with NSP cells. SP cells overexpressed stem cell markers CD133 and ALDH1, pluripotency maintaining factors Nanog, Sox2 and Oct4, oncogenic transcription factor c-Myc, signaling molecule Notch1, and drug resistant gene ABCG2. Moreover, SP cells consistently demonstrated significantly greater tumorigenicity than NSP cells in xenograft model of nude mice. CdODN–SOC efficiently suppressed all CSC properties and phenotypes, and minimized the tumorigenic capability of the SP cells and the resistance to chemotherapy. By comparison, the negative control failed to do so. Conclusion The findings indicate that targeting the key genes conferring the stemness of CSCs can efficiently eliminate CSC-like phenotypes, and thus may be considered a new approach for cancer therapy. Specifically, the present study establishes the combination of Sox2/Oct4/c-Myc targeting as a potential anti-pancreatic cancer agent worthy of further studies in preclinical settings.
Collapse
|
39
|
Asuthkar S, Stepanova V, Lebedeva T, Holterman AL, Estes N, Cines DB, Rao JS, Gondi CS. Multifunctional roles of urokinase plasminogen activator (uPA) in cancer stemness and chemoresistance of pancreatic cancer. Mol Biol Cell 2013; 24:2620-32. [PMID: 23864708 PMCID: PMC3756915 DOI: 10.1091/mbc.e12-04-0306] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is almost always lethal. One of the underlying reasons for this lethality is believed to be the presence of cancer stem cells (CSC), which impart chemoresistance and promote recurrence, but the mechanisms responsible are unclear. Recently the poor prognosis of PDAC has been correlated with increased expression of urokinase plasminogen activator (uPA). In the present study we examine the role of uPA in the generation of PDAC CSC. We observe a subset of cells identifiable as a side population (SP) when sorted by flow cytometry of MIA PaCa-2 and PANC-1 pancreatic cancer cells that possess the properties of CSC. A large fraction of these SP cells are CD44 and CD24 positive, are gemcitabine resistant, possess sphere-forming ability, and exhibit increased tumorigenicity, known characteristics of cancer stemness. Increased tumorigenicity and gemcitabine resistance decrease after suppression of uPA. We observe that uPA interacts directly with transcription factors LIM homeobox-2 (Lhx2), homeobox transcription factor A5 (HOXA5), and Hey to possibly promote cancer stemness. uPA regulates Lhx2 expression by suppressing expression of miR-124 and p53 expression by repressing its promoter by inactivating HOXA5. These results demonstrate that regulation of gene transcription by uPA contributes to cancer stemness and clinical lethality.
Collapse
Affiliation(s)
- Swapna Asuthkar
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL 61605, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Jiang JX, Zhan L, Huang Y, He YZ, Sun CY. SiRNA-mediated silencing of KAP-1 expression inhibits invasion of human pancreatic cancer PANC-1 cells. Shijie Huaren Xiaohua Zazhi 2013; 21:1859-1865. [DOI: 10.11569/wcjd.v21.i19.1859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the impact of small interfering RNA (siRNA)-mediated KAP-1 gene silencing on invasion of human pancreatic cancer PANC-1 cells, and to explore the feasibility of human KAP-1 gene as a therapeutic target for pancreatic cancer.
METHODS: Five KAP-1-specific siRNAs were designed using online software to construct pGC-LV-siRNA-KAP-1 plasmid. Western blot was used to screen the efficient plasmid in 293T cells, and the most efficient one was packed into the recombinant lentivirus Lv-siRNA-KAP-1 in 293T cells. The titer of lentivirus was determined by hole-by-dilution titer assay. The silencing effect of Lv-siRNA-KAP-1 in PANC-1 cells was validated by real-time PCR. After PANC-1 cells were infected with Lv-siRNA-KAP-1, cell invasion was detected by Transwell chamber assay. Vimentin expression in cells was detected by Western blot.
RESULTS: Four KAP-1-specific siRNAs could silence the expression of KAP-1 at the protein level 48 h after transfection. The most efficient pGC-siRNA-3 plasmid was used to construct Lv-VIM-shRNA. The titer of lentivirus was 2 × 109 TU/mL. KAP-1 knockdown significantly inhibited invasion of human pancreatic cancer PANC-1 cells compared to blank control and negative control cells (97.3 ± 25.6 vs 253.3 ± 20.6, 213.2 ± 19.4, both P < 0.05). The expression of vimentin was down-regulated in PANC-1 cells infected with Lv-siRNA-KAP-1.
CONCLUSION: Lv-siRNA-KAP-1 could effectively inhibit the expression of KAP-1 gene in PANC-1 cells in vitro, cell invasion and Vimentin expression. KAP-1 might serve as a new target for gene therapy of pancreatic cancer.
Collapse
|
41
|
Singh S, Chitkara D, Kumar V, Behrman SW, Mahato RI. miRNA profiling in pancreatic cancer and restoration of chemosensitivity. Cancer Lett 2013; 334:211-20. [DOI: 10.1016/j.canlet.2012.10.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Revised: 09/26/2012] [Accepted: 10/07/2012] [Indexed: 12/13/2022]
|
42
|
Jiang JX, Gao S, Wang M, Li X, Sun CY. Identification and bioinformatic analysis of differentially expressed genes in pancreatic cancer stem cells. Shijie Huaren Xiaohua Zazhi 2013; 21:1218-1225. [DOI: 10.11569/wcjd.v21.i13.1218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To identify differentially expressed genes in pancreatic cancer stem cells and to analyze them by bioinformatics.
METHODS: MIA-PaCa2 (TIChigh) and BxPc-3 (TIClow) were used as tool cells in this study. Total RNA was prepared from MIA-PaCa2 (TIChigh) and BxPc-3 (TIClow) cells. After quality identification and fluorescent labeling, RNA samples were hybridized with the Agilent human gene expression microarrays. Raw data were normalized using Quantile algorithm and Gene Spring Software 11.0. Gene Ontology database and KEGG database were used for GO and Pathway comments on differentially expressed genes, respectively.
RESULTS: A total of 7059 differentially expressed genes were identified (P < 0.05, fold change ≥ 2), including 3440 up-regulated and 3619 down-regulated genes in MIA-PaCa2 (TIChigh) cells. These differentially expressed genes were mainly involved in polyamine metabolic process, nucleobase-containing small molecule interconversion, histone H4 acetylation, cell division, cell cycle, apoptotic process, and toll-like receptor signaling pathway. GO analysis demonstrated they are involved in cancer, endocytosis, O-glycan biosynthesis, pyrimidine metabolism, and glutathione metabolism. Pathway analysis showed the presence of crosstalk among several signaling pathways.
CONCLUSION: Differentially expressed genes identified in this study may be involved in the regulation of different signaling pathways and the genesis of pancreatic cancer stem cells. They might become new therapeutic targets for pancreatic cancer.
Collapse
|
43
|
Jiang JX, Zhan L, Huang Y, He YZ, Sun CY. Clinical significance of expression of KAP-1 in pancreatic carcinoma. Shijie Huaren Xiaohua Zazhi 2013; 21:829-834. [DOI: 10.11569/wcjd.v21.i9.829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression of KAP-1 in pancreatic carcinoma.
METHODS: The expression of KAP-1 protein in 46 pancreatic cancer specimens (including 15 cases of highly differentiated cancer, 17 cases of moderately differentiated cancer, and 14 cases of poorly differentiated cancer) and 9 normal pancreas specimens was detected by immunohistochemistry. The mRNA and protein expression of KAP-1 in 8 cases of pancreatic cancer and matched tumor-adjacent pancreatic tissue and in pancreatic carcinoma cell lines (BxPC3, CAPAN-1, PANC-1, AsPC-1, SW1990, MiaPaCa-2 and CFPAC-1) was also detected by RT-qPCR and Western blot.
RESULTS: The positive rate of KAP-1 expression was 45.6% (21/46) in pancreatic cancer and 11.1% (1/9) in normal pancreatic tissue, and was 78.6% (11/14) in poorly differentiated pancreatic cancer, 47.1% (8/17) in moderately differentiated pancreatic cancer, and 13.3% (2/15) in highly differentiated pancreatic cancer. The mRNA and protein expression of KAP-1 was higher in pancreatic cancer than in matched tumor-adjacent pancreatic tissue. The mRNA expression of KAP-1 was highest in poorly differentiated pancreatic cancer line Panc-1, higher in BXPC-3 and CFPAC-1, lower in SW1990, Capan-1 and MIAPaCa-2, and lowest in AsPC-1 and Capan-2. The protein expression of KAP-1 was highest in poorly differentiated pancreatic cancer line MIAPaCa-2 and Panc-1, higher in CFPAC-1 which is derived from liver metastases of pancreatic cancer cell, and was undetectable in other cell lines.
CONCLUSION: The expression of KAP-1 in human pancreatic cancer tissue is significantly higher than that in normal pancreatic tissue, and KAP-1 expression is involved in pancreatic cancer cell differentiation. KAP-1 may play an important role in the development of pancreatic cancer.
Collapse
|
44
|
Jiang JX, Gao S, Pan YZ, Sun CY. Quantitative proteomic analysis of differentially expressed proteins in pancreatic cancer stem cells. Shijie Huaren Xiaohua Zazhi 2013; 21:145-152. [DOI: 10.11569/wcjd.v21.i2.145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To screen and identify differentially expressed proteins in pancreatic cancer stem cells.
METHODS: MIA-PaCa2 (TIChigh) and BxPc-3 (TIClow) were used in the study. Differentially expressed proteins between MIA-PaCa2 (TIChigh) and BxPc-3 (TIClow) cells were isolated and screened by 2D-DIGE analysis. Protein identification was performed by peptide mass fingerprinting with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF/TOF). Western blot was performed to verify the differential expression of TRIM28.
RESULTS: Fluorescent differential protein expression patterns were obtained between MIA-PaCa2 (TIChigh) and BxPc-3 (TIClow) cells. Analyses with DeCyder v6.5 software showed a total of 23 differentially expressed protein spots (>1.5 folds), and these protein spots were identified by mass spectrometry as 19 proteins, which are involved in cell communication and signal transduction, immune response, transcription and cell cycle regulation, adipocyte differentiation and lipid droplet formation, cytoskeletal formation, cell adhesion, transport, and translation. Western blot analysis revealed that TRIM28 was highly expressed in MIA-PaCa2 (TIChigh) cells but not expressed in BxPc-3 (TIClow) cells. Among the 19 identified proteins, 8 were up-regulated and 11 down-regulated in MIA-PaCa2 (TIChigh) cells.
CONCLUSION: The identified differentially expressed proteins, such as TRIM28, are associated with the genesis, development and regulation of pancreatic cancer stem cells. They may become new therapeutic targets for pancreatic cancer.
Collapse
|
45
|
Jiang JX, Gao S, Wang M, Li X, Sun CY. Identification of differentially expressed microRNAs in pancreatic cancer stem cells. Shijie Huaren Xiaohua Zazhi 2012; 20:3354-3360. [DOI: 10.11569/wcjd.v20.i34.3354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To screen and identify differentially expressed miRNAs in pancreatic cancer stem cells and to analyze them by bioinformatics.
METHODS: Total RNA was prepared from MIA-PaCa2 (TIChigh) and BxPc-3 (TIClow) cells. After quality identification and fluorescent labeling, the RNA samples were hybridized with Agilent human miRNA microarrays. Raw data were normalized using Quantile algorithm and Gene Spring Software 11.0. Part of differentially expressed miRNAs were validated by SYBR Green real-time PCR. Sanger database was used to predict target genes for differentially expressed miRNAs. Gene Ontology database and KEGG database were used for GO annotations and Pathway annotations of target genes, respectively.
RESULTS: A total of 940 miRNAs were identified and among them 91 were differentially expressed (P < 0.05, fold change ≥ 2), including 45 up-regulated and 46 down-regulated ones in MIA-PaCa2 (TIChigh). Twenty-one miRNAs were validated by real-time PCR, and the results for 19 miRNAs were consistent with those of chip detection. 2 895 target genes were predicted for 91 differentially expressed miRNAs, and these genes were mainly involved in events, such as axon guidance, angiogenesis, and post-translational protein modification, and in pathways such as cancer, focal adhesion, Hedgehog signaling pathway, and MAPK signaling pathway.
CONCLUSION: Many differentially expressed miRNAs in pancreatic cancer stem cells have been identified, and their target genes are involved in a variety of biological events and in multiple signal pathways. These differentially expressed miRNAs may become new therapeutic targets for pancreatic cancer.
Collapse
|
46
|
Tang J, Cai H, Lin L, Xie P, Zhong W, Tang M. Increased expression of CD24 is associated with tumor progression and prognosis in patients suffering osteosarcoma. Clin Transl Oncol 2012; 15:541-7. [PMID: 23143956 DOI: 10.1007/s12094-012-0961-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 10/10/2012] [Indexed: 01/06/2023]
Abstract
OBJECTIVE As a small heavily glycosylated mucin-like glycosyl-phosphatidylinositol-anchored cell surface protein, CD24 plays an important role in carcinogenesis of various human malignancies. However, its involvement in osteosarcoma is still unclear. The aim of this study was to investigate the expression pattern and the clinical significance of CD24 in human osteosarcoma. METHODS CD24 mRNA and protein expression levels were, respectively, detected by RT-PCR and Western blot assays using 30 pairs of osteosarcoma and noncancerous bone tissues. Then, immunohistochemistry was performed to analyze the association of CD24 expression in 166 osteosarcoma tissues with clinicopathological factors or survival of patients. RESULTS CD24 expression at mRNA and protein levels were both significantly higher in osteosarcoma tissues than those in corresponding noncancerous bone tissues (both P < 0.001). In addition, CD24 protein was positively expressed in 129 of 166 (77.7 %) osteosarcoma specimens with a cytoplasmic and membraneous staining, and also increased in the osteosarcoma specimens with advanced clinical stage (P = 0.01) and positive distant metastasis (P = 0.005). The univariate and multivariate analyses showed that osteosarcoma patients with high CD24 expression had poorer overall and disease-free survival, and high CD24 expression was an independent prognostic factor for both overall and disease-free survival. CONCLUSION The aforementioned findings offer convincing evidence for the first time that the increased expression of CD24 is correlated with tumor aggressiveness and tumor metastasis of osteosarcoma, and this molecule is an independent prognostic marker for osteosarcoma patients.
Collapse
Affiliation(s)
- J Tang
- Orthopedics Department, Xuhui Central Hospital, No. 966, Middle Huaihai Road, Shanghai, 200031, China
| | | | | | | | | | | |
Collapse
|
47
|
Kim MS, Kuppireddy SV, Sakamuri S, Singal M, Getnet D, Harsha HC, Goel R, Balakrishnan L, Jacob HKC, Kashyap MK, Tankala SG, Maitra A, Iacobuzio-Donahue CA, Jaffee E, Goggins MG, Velculescu VE, Hruban RH, Pandey A. Rapid characterization of candidate biomarkers for pancreatic cancer using cell microarrays (CMAs). J Proteome Res 2012; 11:5556-5563. [PMID: 22985314 PMCID: PMC3565537 DOI: 10.1021/pr300483r] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tissue microarrays have become a valuable tool for high-throughput analysis using immunohistochemical labeling. However, the large majority of biochemical studies are carried out in cell lines to further characterize candidate biomarkers or therapeutic targets with subsequent studies in animals or using primary tissues. Thus, cell line-based microarrays could be a useful screening tool in some situations. Here, we constructed a cell microarray (CMA) containing a panel of 40 pancreatic cancer cell lines available from American Type Culture Collection in addition to those locally available at Johns Hopkins. As proof of principle, we performed immunocytochemical labeling of an epithelial cell adhesion molecule (Ep-CAM), a molecule generally expressed in the epithelium, on this pancreatic cancer CMA. In addition, selected molecules that have been previously shown to be differentially expressed in pancreatic cancer in the literature were validated. For example, we observed strong labeling of CA19-9 antigen, a prognostic and predictive marker for pancreatic cancer. We also carried out a bioinformatics analysis of a literature curated catalog of pancreatic cancer biomarkers developed previously by our group and identified two candidate biomarkers, HLA class I and transmembrane protease, serine 4 (TMPRSS4), and examined their expression in the cell lines represented on the pancreatic cancer CMAs. Our results demonstrate the utility of CMAs as a useful resource for rapid screening of molecules of interest and suggest that CMAs can become a universal standard platform in cancer research.
Collapse
Affiliation(s)
- Min-Sik Kim
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Sarada V. Kuppireddy
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Sruthi Sakamuri
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Mukul Singal
- Government Medical College and Hospital, Chandigarh 160030, India
| | - Derese Getnet
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - H. C. Harsha
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
| | - Renu Goel
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
| | - Lavanya Balakrishnan
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
| | - Harrys K. C. Jacob
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
| | - Manoj K. Kashyap
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
| | | | - Anirban Maitra
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Pathology Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland 21231, United States
| | - Christine A. Iacobuzio-Donahue
- Department of Pathology Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland 21231, United States
| | - Elizabeth Jaffee
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland 21231, United States
| | - Michael G. Goggins
- Department of Pathology Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland 21231, United States
| | - Victor E. Velculescu
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins Kimmel Cancer Center, Baltimore, Maryland 21231, United States
| | - Ralph H. Hruban
- Department of Pathology Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland 21231, United States
| | - Akhilesh Pandey
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Pathology Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland 21231, United States
| |
Collapse
|
48
|
Ghosh SC, Neslihan Alpay S, Klostergaard J. CD44: a validated target for improved delivery of cancer therapeutics. Expert Opin Ther Targets 2012; 16:635-50. [PMID: 22621669 DOI: 10.1517/14728222.2012.687374] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Advances in cancer therapeutics, namely more effective and less toxic treatments, will occur with targeting strategies that enhance the tumor biodistribution and thwart normal tissue exposure of the drug. This review focuses on cancer drug targeting approaches that exploit the expression of the cell-surface proteoglycan family, CD44, on the tumor cell surface followed by some form of ligand binding and induced CD44 internalization and intracellular drug release: in effect using this as a 'Trojan Horse' to more selectively access tumor cells. AREAS COVERED This review defines the origins of evidence for a linkage between CD44 expression and malignancy, and invokes contemporary views of the importance of putative CD44(+) cancer stem cells in disease resistance. Although the primary emphasis is on the most advanced and developed paths, those that have either made it to the clinic or are well-poised to get there, a wide scope of additional approaches at various preclinical stages is also briefly reviewed. EXPERT OPINION The future should see development of drug targeting approaches that exploit CD44 expression on CSCs/TICs, including applications to cytotoxic agents currently in the clinic.
Collapse
Affiliation(s)
- Sukhen C Ghosh
- The University of Texas, Institute of Molecular Medicine, Health Science Center, Center for Molecular Imaging, 1825 Pressler Street, SRB 330C, Houston, TX 77030, USA
| | | | | |
Collapse
|