1
|
Macrophage Inhibitory Factor-1 (MIF-1) controls the plasticity of multiple myeloma tumor cells. PLoS One 2018; 13:e0206368. [PMID: 30383785 PMCID: PMC6211687 DOI: 10.1371/journal.pone.0206368] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/11/2018] [Indexed: 11/29/2022] Open
Abstract
Multiple Myeloma (MM) is the second most common hematological malignancy with a median survival of 5–10 years. While current treatments initially cause remission, relapse almost always occurs, leading to the hypothesis that a chemotherapy-resistant cancer stem cell (CSC) remains dormant, and undergoes self-renewal and differentiation to reestablish disease. Our finding is that the mature cancer cell (CD138+, rapidly proliferating and chemosensitive) has developmental plasticity; namely, the ability to dedifferentiate back into its own chemoresistant CSC progenitor, the CD138–, quiescent pre-plasma cell. We observe multiple cycles of differentiation and dedifferentiation in the absence of niche or supportive accessory cells, suggesting that soluble cytokines secreted by the MM cells themselves are responsible for this bidirectional interconversion and that stemness and chemoresistance are dynamic characteristics that can be acquired or lost and thus may be targetable. By examining cytokine secretion of CD138- and CD138+ RPMI-8226 cells, we identified that concomitant with interconversion, Macrophage Migration Inhibitory Factor (MIF-1) is secreted. The addition of a small molecule MIF-1 inhibitor (4-IPP) or MIF-1 neutralizing antibodies to CD138+ cells accelerated dedifferentiation back into the CD138- progenitor, while addition of recombinant MIF-1 drove cells towards CD138+ differentiation. A similar increase in the CD138- population is seen when MM tumor cells isolated from primary bone marrow aspirates are cultured in the presence of 4-IPP. As the CD138+ MM cell is chemosensitive, targeting MIF-1 and/or the pathways that it regulates could be a viable way to modulate stemness and chemosensitivity, which could in turn transform the treatment of MM.
Collapse
|
2
|
The p53 Pathway in Glioblastoma. Cancers (Basel) 2018; 10:cancers10090297. [PMID: 30200436 PMCID: PMC6162501 DOI: 10.3390/cancers10090297] [Citation(s) in RCA: 266] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/17/2018] [Accepted: 08/28/2018] [Indexed: 12/27/2022] Open
Abstract
The tumor suppressor and transcription factor p53 plays critical roles in tumor prevention by orchestrating a wide variety of cellular responses, including damaged cell apoptosis, maintenance of genomic stability, inhibition of angiogenesis, and regulation of cell metabolism and tumor microenvironment. TP53 is one of the most commonly deregulated genes in cancer. The p53-ARF-MDM2 pathway is deregulated in 84% of glioblastoma (GBM) patients and 94% of GBM cell lines. Deregulated p53 pathway components have been implicated in GBM cell invasion, migration, proliferation, evasion of apoptosis, and cancer cell stemness. These pathway components are also regulated by various microRNAs and long non-coding RNAs. TP53 mutations in GBM are mostly point mutations that lead to a high expression of a gain of function (GOF) oncogenic variants of the p53 protein. These relatively understudied GOF p53 mutants promote GBM malignancy, possibly by acting as transcription factors on a set of genes other than those regulated by wild type p53. Their expression correlates with worse prognosis, highlighting their potential importance as markers and targets for GBM therapy. Understanding mutant p53 functions led to the development of novel approaches to restore p53 activity or promote mutant p53 degradation for future GBM therapies.
Collapse
|
3
|
Balandrán JC, Purizaca J, Enciso J, Dozal D, Sandoval A, Jiménez-Hernández E, Alemán-Lazarini L, Perez-Koldenkova V, Quintela-Núñez Del Prado H, Rios de Los Ríos J, Mayani H, Ortiz-Navarrete V, Guzman ML, Pelayo R. Pro-inflammatory-Related Loss of CXCL12 Niche Promotes Acute Lymphoblastic Leukemic Progression at the Expense of Normal Lymphopoiesis. Front Immunol 2017; 7:666. [PMID: 28111575 PMCID: PMC5216624 DOI: 10.3389/fimmu.2016.00666] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 12/19/2016] [Indexed: 01/04/2023] Open
Abstract
Pediatric oncology, notably childhood acute lymphoblastic leukemia (ALL), is currently one of the health-leading concerns worldwide and a biomedical priority. Decreasing overall leukemia mortality in children requires a comprehensive understanding of its pathobiology. It is becoming clear that malignant cell-to-niche intercommunication and microenvironmental signals that control early cell fate decisions are critical for tumor progression. We show here that the mesenchymal stromal cell component of ALL bone marrow (BM) differ from its normal counterpart in a number of functional properties and may have a key role during leukemic development. A decreased proliferation potential, contrasting with the strong ability of producing pro-inflammatory cytokines and an aberrantly loss of CXCL12 and SCF, suggest that leukemic lymphoid niches in ALL BM are unique and may exclude normal hematopoiesis. Cell competence ex vivo assays within tridimensional coculture structures indicated a growth advantage of leukemic precursor cells and their niche remodeling ability by CXCL12 reduction, resulting in leukemic cell progression at the expense of normal niche-associated lymphopoiesis.
Collapse
Affiliation(s)
- Juan Carlos Balandrán
- Oncology Research Unit, Mexican Institute for Social Security, Mexico City, Mexico; Molecular Biomedicine Program, CINVESTAV, IPN, Mexico City, Mexico
| | - Jessica Purizaca
- Oncology Research Unit, Mexican Institute for Social Security , Mexico City , Mexico
| | - Jennifer Enciso
- Oncology Research Unit, Mexican Institute for Social Security, Mexico City, Mexico; Biochemistry Sciences Program, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico
| | - David Dozal
- Hospital para el Niño, Instituto Materno Infantil del Estado de México , Toluca , México
| | - Antonio Sandoval
- Hospital para el Niño, Instituto Materno Infantil del Estado de México , Toluca , México
| | | | | | - Vadim Perez-Koldenkova
- Laboratorio de Microscopía, Centro de Instrumentos, Coordinación de Investigación en Salud, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social , Mexico City , México
| | | | - Jussara Rios de Los Ríos
- Oncology Research Unit, Mexican Institute for Social Security, Mexico City, Mexico; Biochemistry Sciences Program, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico
| | - Héctor Mayani
- Oncology Research Unit, Mexican Institute for Social Security , Mexico City , Mexico
| | | | - Monica L Guzman
- Division of Hematology and Medical Oncology, Weill Cornell Medicine , New York, NY , USA
| | - Rosana Pelayo
- Oncology Research Unit, Mexican Institute for Social Security , Mexico City , Mexico
| |
Collapse
|
4
|
Bayir E, Bilgi E, Urkmez AS. Implementation of Nanoparticles in Cancer Therapy. PHARMACEUTICAL SCIENCES 2017. [DOI: 10.4018/978-1-5225-1762-7.ch047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Cancer is a wide group of diseases and generally characterized by uncontrolled proliferation of cells whose metabolic activities are disrupted. Conventionally, chemotherapy, radiotherapy, and surgery are used in the treatment of cancer. However, in theory, even a single cancer cell may trigger recurrence. Therefore, these treatments cannot provide high survival rate for deadly types. Identification of alternative methods in treatment of cancers is inevitable because of adverse effects of conventional methods. In the last few decades, nanotechnology developed by scientists working in different disciplines—physics, chemistry, and biology—offers great opportunities. It is providing elimination of both circulating tumor cells and solid cancer cells by targeting cancer cells. In this chapter, inadequate parts of conventional treatment methods, nanoparticle types used in new treatment methods of cancer, and targeting methods of nanoparticles are summarized; furthermore, recommendations of future are provided.
Collapse
|
5
|
Vega SL, Liu E, Arvind V, Bushman J, Sung HJ, Becker ML, Lelièvre S, Kohn J, Vidi PA, Moghe PV. High-content image informatics of the structural nuclear protein NuMA parses trajectories for stem/progenitor cell lineages and oncogenic transformation. Exp Cell Res 2016; 351:11-23. [PMID: 28034673 DOI: 10.1016/j.yexcr.2016.12.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/20/2016] [Accepted: 12/22/2016] [Indexed: 12/15/2022]
Abstract
Stem and progenitor cells that exhibit significant regenerative potential and critical roles in cancer initiation and progression remain difficult to characterize. Cell fates are determined by reciprocal signaling between the cell microenvironment and the nucleus; hence parameters derived from nuclear remodeling are ideal candidates for stem/progenitor cell characterization. Here we applied high-content, single cell analysis of nuclear shape and organization to examine stem and progenitor cells destined to distinct differentiation endpoints, yet undistinguishable by conventional methods. Nuclear descriptors defined through image informatics classified mesenchymal stem cells poised to either adipogenic or osteogenic differentiation, and oligodendrocyte precursors isolated from different regions of the brain and destined to distinct astrocyte subtypes. Nuclear descriptors also revealed early changes in stem cells after chemical oncogenesis, allowing the identification of a class of cancer-mitigating biomaterials. To capture the metrology of nuclear changes, we developed a simple and quantitative "imaging-derived" parsing index, which reflects the dynamic evolution of the high-dimensional space of nuclear organizational features. A comparative analysis of parsing outcomes via either nuclear shape or textural metrics of the nuclear structural protein NuMA indicates the nuclear shape alone is a weak phenotypic predictor. In contrast, variations in the NuMA organization parsed emergent cell phenotypes and discerned emergent stages of stem cell transformation, supporting a prognosticating role for this protein in the outcomes of nuclear functions.
Collapse
Affiliation(s)
- Sebastián L Vega
- Department of Chemical and Biochemical Engineering, Rutgers University, Piscataway, NJ, United States
| | - Er Liu
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, United States
| | - Varun Arvind
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, United States
| | - Jared Bushman
- Department of Chemistry and Chemical Biology, New Jersey Center for Biomaterials, Piscataway, NJ, United States; School of Pharmacy, University of Wyoming, Laramie, WY, United States
| | - Hak-Joon Sung
- Department of Chemistry and Chemical Biology, New Jersey Center for Biomaterials, Piscataway, NJ, United States; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
| | - Matthew L Becker
- Department of Polymer Science and Engineering, University of Akron, Akron, OH, United States
| | - Sophie Lelièvre
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States
| | - Joachim Kohn
- Department of Chemistry and Chemical Biology, New Jersey Center for Biomaterials, Piscataway, NJ, United States
| | - Pierre-Alexandre Vidi
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, United States.
| | - Prabhas V Moghe
- Department of Chemical and Biochemical Engineering, Rutgers University, Piscataway, NJ, United States; Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, United States.
| |
Collapse
|
6
|
de la Puente P, Quan N, Hoo RS, Muz B, Gilson RC, Luderer M, King J, Achilefu S, Salama NN, Vij R, Azab AK. Newly established myeloma-derived stromal cell line MSP-1 supports multiple myeloma proliferation, migration, and adhesion and induces drug resistance more than normal-derived stroma. Haematologica 2016; 101:e307-11. [PMID: 27081175 DOI: 10.3324/haematol.2016.142190] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Pilar de la Puente
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, MO, USA
| | - Nancy Quan
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, MO, USA Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, MO, USA
| | - Ryan Soo Hoo
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, MO, USA Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, MO, USA
| | - Barbara Muz
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, MO, USA
| | - Rebecca C Gilson
- Biomedical Engineering, Washington University in Saint Louis School of Medicine, MO, USA
| | - Micah Luderer
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, MO, USA
| | - Justin King
- Section of Stem Cell Transplant and Leukemia, Division of Medical Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Samuel Achilefu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Noha Nabil Salama
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, MO, USA Department of Pharmaceutics and Industrial Pharmacy, University Faculty of Pharmacy, Cairo, Egypt
| | - Ravi Vij
- Section of Stem Cell Transplant and Leukemia, Division of Medical Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, MO, USA
| |
Collapse
|
7
|
de la Puente P, Azab AK. 3D tissue-engineered bone marrow: what does this mean for the treatment of multiple myeloma? Future Oncol 2016; 12:1545-7. [PMID: 27333447 DOI: 10.2217/fon-2016-0057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Pilar de la Puente
- Cancer Biology Division, Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, St Louis, MO, USA
| | - Abdel Kareem Azab
- Cancer Biology Division, Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, St Louis, MO, USA
| |
Collapse
|
8
|
Chen S, Li H, Li S, Yu J, Wang M, Xing H, Tang K, Tian Z, Rao Q, Wang J. Rac1 GTPase Promotes Interaction of Hematopoietic Stem/Progenitor Cell with Niche and Participates in Leukemia Initiation and Maintenance in Mouse. Stem Cells 2016; 34:1730-41. [PMID: 26946078 DOI: 10.1002/stem.2348] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 01/17/2016] [Accepted: 02/10/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Shuying Chen
- State Key Laboratory of Experimental Hematology; Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin P. R. China
| | - Huan Li
- State Key Laboratory of Experimental Hematology; Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin P. R. China
| | - Shouyun Li
- State Key Laboratory of Experimental Hematology; Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin P. R. China
| | - Jing Yu
- State Key Laboratory of Experimental Hematology; Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin P. R. China
| | - Min Wang
- State Key Laboratory of Experimental Hematology; Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin P. R. China
| | - Haiyan Xing
- State Key Laboratory of Experimental Hematology; Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin P. R. China
| | - Kejing Tang
- State Key Laboratory of Experimental Hematology; Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin P. R. China
| | - Zheng Tian
- State Key Laboratory of Experimental Hematology; Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin P. R. China
| | - Qing Rao
- State Key Laboratory of Experimental Hematology; Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin P. R. China
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology; Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin P. R. China
| |
Collapse
|
9
|
Molaeipour Z, Shamsasanjan K, Movassaghpour AA, Akbarzadehlaleh P, Sabaghi F, Saleh M. The Effect of Bone Marrow Mesenchymal Stem Cells on Vitamin D3 Induced Monocytic Differentiation of U937 Cells. Adv Pharm Bull 2016; 6:23-9. [PMID: 27123414 DOI: 10.15171/apb.2016.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/27/2015] [Accepted: 01/07/2016] [Indexed: 12/30/2022] Open
Abstract
PURPOSE Mesenchymal stem cells (MSCs) are key components of the hematopoietic stem cells (HSCs) niche. They control the process of hematopoiesis by secreting regulatory cytokines, growth factors and expression of important cell adhesion molecules for cell-tocell interactions. In this research, we have investigated the effect of bone marrow derived MSCs on monocytic differentiation of U937 cells line. METHODS U937 cells were cultured in both direct co-culture with MSCs and MSCs conditioned medium (C.M) driven. This study used 1,25-dihydroxyvitamin D3(VitD3) as inductor of monocytic differentiation and U937 cells treated with VitD3 morphology was examined by Wright Giemsa staining. CD14 monocytic differentiation marker was measured by flow cytometry and monocytic gene expression was assessed by real time polymerase chain reaction (RT PCR). RESULTS The results of flow cytometric analysis showed that CD14 expression of U937 increased. The higher effect of MSCs co-culture on CD14 expression in U937 cells was observed, compared to the conditioned medium. Among ten monocytic related genes which were screened that was observed increase in 5 genes in which CXCR4 and CSF2RA showed significant increase. CONCLUSION The results obtained show that MSCs have supportive effect on the monocytic differentiation of U937 cells. However, a distinct mechanism of that remains unclear.
Collapse
Affiliation(s)
- Zahra Molaeipour
- Hematology Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasanjan
- Hematology Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Parvin Akbarzadehlaleh
- Pharmaceutical Biotechnology Department, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Sabaghi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mahshid Saleh
- Hematology Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
10
|
Molaeipour Z, Shamsasanjan K, Movassaghpour AA, Akbarzadehlaleh P, Sabaghi F, Saleh M. The Effect of Bone Marrow Mesenchymal Stem Cells on Vitamin D3 Induced Monocytic Differentiation of U937 Cells. Adv Pharm Bull 2016; 6:23-29. [PMID: 27123414 PMCID: PMC4845548 DOI: 10.15171/apb.2016.05] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/27/2015] [Accepted: 01/07/2016] [Indexed: 08/05/2023] Open
Abstract
PURPOSE Mesenchymal stem cells (MSCs) are key components of the hematopoietic stem cells (HSCs) niche. They control the process of hematopoiesis by secreting regulatory cytokines, growth factors and expression of important cell adhesion molecules for cell-tocell interactions. In this research, we have investigated the effect of bone marrow derived MSCs on monocytic differentiation of U937 cells line. METHODS U937 cells were cultured in both direct co-culture with MSCs and MSCs conditioned medium (C.M) driven. This study used 1,25-dihydroxyvitamin D3(VitD3) as inductor of monocytic differentiation and U937 cells treated with VitD3 morphology was examined by Wright Giemsa staining. CD14 monocytic differentiation marker was measured by flow cytometry and monocytic gene expression was assessed by real time polymerase chain reaction (RT PCR). RESULTS The results of flow cytometric analysis showed that CD14 expression of U937 increased. The higher effect of MSCs co-culture on CD14 expression in U937 cells was observed, compared to the conditioned medium. Among ten monocytic related genes which were screened that was observed increase in 5 genes in which CXCR4 and CSF2RA showed significant increase. CONCLUSION The results obtained show that MSCs have supportive effect on the monocytic differentiation of U937 cells. However, a distinct mechanism of that remains unclear.
Collapse
Affiliation(s)
- Zahra Molaeipour
- Hematology Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasanjan
- Hematology Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Parvin Akbarzadehlaleh
- Pharmaceutical Biotechnology Department, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Sabaghi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mahshid Saleh
- Hematology Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
11
|
de la Puente P, Muz B, Gilson RC, Azab F, Luderer M, King J, Achilefu S, Vij R, Azab AK. 3D tissue-engineered bone marrow as a novel model to study pathophysiology and drug resistance in multiple myeloma. Biomaterials 2015; 73:70-84. [PMID: 26402156 PMCID: PMC4917006 DOI: 10.1016/j.biomaterials.2015.09.017] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/08/2015] [Accepted: 09/10/2015] [Indexed: 01/03/2023]
Abstract
PURPOSE Multiple myeloma (MM) is the second most prevalent hematological malignancy and it remains incurable despite the introduction of several novel drugs. The discrepancy between preclinical and clinical outcomes can be attributed to the failure of classic two-dimensional (2D) culture models to accurately recapitulate the complex biology of MM and drug responses observed in patients. EXPERIMENTAL DESIGN We developed 3D tissue engineered bone marrow (3DTEBM) cultures derived from the BM supernatant of MM patients to incorporate different BM components including MM cells, stromal cells, and endothelial cells. Distribution and growth were analyzed by confocal imaging, and cell proliferation of cell lines and primary MM cells was tested by flow cytometry. Oxygen and drug gradients were evaluated by immunohistochemistry and flow cytometry, and drug resistance was studied by flow cytometry. RESULTS 3DTEBM cultures allowed proliferation of MM cells, recapitulated their interaction with the microenvironment, recreated 3D aspects observed in the bone marrow niche (such as oxygen and drug gradients), and induced drug resistance in MM cells more than 2D or commercial 3D tissue culture systems. CONCLUSIONS 3DTEBM cultures not only provide a better model for investigating the pathophysiology of MM, but also serve as a tool for drug development and screening in MM. In the future, we will use the 3DTEBM cultures for developing personalized therapeutic strategies for individual MM patients.
Collapse
Affiliation(s)
- Pilar de la Puente
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Barbara Muz
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Rebecca C Gilson
- Biomedical Engineering, Washington University School of Medicine, St. Louis, MO, USA
| | - Feda Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Micah Luderer
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Justin King
- Section of Stem Cell Transplant and Leukemia, Division of Medical Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Samuel Achilefu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ravi Vij
- Section of Stem Cell Transplant and Leukemia, Division of Medical Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
12
|
Smith J, Smith A, Shelton R, Cooper P. Dental Pulp Cell Behavior in Biomimetic Environments. J Dent Res 2015; 94:1552-9. [DOI: 10.1177/0022034515599767] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
There is emerging recognition of the importance of a physiologically relevant in vitro cell culture environment to promote maintenance of stem cells for tissue engineering and regenerative medicine purposes. In vivo, appropriate cellular cues are provided by local tissue extracellular matrix (ECM), and these are not currently recapitulated well in vitro using traditional cultureware. We therefore hypothesized that better replication of the in vivo environment for cell culture and differentiation could be achieved by culturing dental pulp cells with their associated ECM. Primary dental pulp cells were subsequently seeded onto pulp-derived ECM-coated cultureware. While at up to 24 h they exhibited the same level of adherence as those cells seeded on tissue culture–treated surfaces, by 4 d cell numbers and proliferation rates were significantly decreased in cells grown on pulp ECM compared with controls. Analysis of stem cell and differentiation marker transcripts, as well as Oct 3/4 protein distribution, supported the hypothesis that cells cultured on ECM better maintained a stem cell phenotype compared with those cultured on standard tissue culture–treated surfaces. Subsequent differentiation analysis of cells cultured on ECM demonstrated that they exhibited enhanced mineralization, as determined by alizarin red staining and mineralized marker expression. Supplementation of a 3% alginate hydrogel with pulp ECM components and dental pulp cells followed by differentiation induction in mineralization medium resulted in a time-dependent mineral deposition at the periphery of the construct, as demonstrated histologically and using micro–computed tomography analysis, which was reminiscent of tooth structure. In conclusion, data indicate that culture of pulp cells in the presence of ECM better replicates the in vivo environment, maintaining a stem cell phenotype suitable for downstream tissue engineering applications.
Collapse
Affiliation(s)
- J.G. Smith
- Oral Biology, School of Dentistry, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - A.J. Smith
- Oral Biology, School of Dentistry, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - R.M. Shelton
- Biomaterials, School of Dentistry, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - P.R. Cooper
- Oral Biology, School of Dentistry, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
13
|
Zhu H, Wang D, Liu Y, Su Z, Zhang L, Chen F, Zhou Y, Wu Y, Yu M, Zhang Z, Shao G. Role of the Hypoxia-inducible factor-1 alpha induced autophagy in the conversion of non-stem pancreatic cancer cells into CD133+ pancreatic cancer stem-like cells. Cancer Cell Int 2013; 13:119. [PMID: 24305593 PMCID: PMC4177138 DOI: 10.1186/1475-2867-13-119] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 11/24/2013] [Indexed: 12/13/2022] Open
Abstract
The initiation and progression of various solid tumors, including pancreatic carcinoma, are driven by a population of cells with stem cell properties, namely cancer stem cells (CSCs). Like their normal counterparts, CSCs are also believed to rely on their own microenvironment termed niches to sustain the population. Hypoxia-inducible factor-1α (HIF-1α) is a major actor in the cell survival response to hypoxia. Recently, several researchers proposed that non-stem cancer cells can convert to stem-like cells to maintain equilibrium. The present study focuses on whether non-stem pancreatic cancer cells can convert to stem-like cells and the role of HIF-1α and autophagy in modulating this conversation. The non-stem pancreatic cancer cells and pancreatic cancer stem-like cells were separated by magnetic sorting column. Intermittent hypoxia enhanced stem-like properties of non-stem pancreatic cancer cells and stimulated the levels of HIF-1α, LC3-II and Beclin. Enhanced autophagy was associated with the elevated level of HIF-1α. The conversation of non-stem pancreatic cancer cells into pancreatic cancer stem-like cells was induced by HIF-1α and autophagy. This novel finding may indicate the specific role of HIF-1α and autophagy in promoting the dynamic equilibrium between CSCs and non-CSCs. Also, it emphasizes the importance of developing therapeutic strategies targeting cancer stem cells as well as the microenvironmental influence on the tumor.
Collapse
Affiliation(s)
| | - Dongqing Wang
- The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Zhu L, Cheng X, Ding Y, Shi J, Jin H, Wang H, Wu Y, Ye J, Lu Y, Wang TC, Yang CS, Tu SP. Bone marrow-derived myofibroblasts promote colon tumorigenesis through the IL-6/JAK2/STAT3 pathway. Cancer Lett 2013; 343:80-9. [PMID: 24145153 DOI: 10.1016/j.canlet.2013.09.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 09/09/2013] [Accepted: 09/14/2013] [Indexed: 12/14/2022]
Abstract
Bone marrow-derived myofibroblasts (BMFs) have been shown to promote tumor growth. Here, we found that BMFs or BMF conditioned medium (BMF-CM) induced cancer stem cell-like sphere formation of colon cancer cells. The co-cultured BMFs, but not co-cultured cancer cells, expressed higher levels of IL-6 than BMFs or cancer cells cultured alone. Anti-mouse IL-6 neutralizing antibody, JAK2 inhibitors and STAT3 knockdown in mouse cancer cells reduced BMF- and BMF-CM-induced sphere formation of colon cancer cells. When co-injected, BMFs significantly enhanced tumorigenesis of colon cancer cells in mice. Our results demonstrate that BMFs promote tumorigenesis via the activation of the IL-6/JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Liming Zhu
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China; Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Xiaojiao Cheng
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Yanfei Ding
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Jindong Shi
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Huanyu Jin
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Hong Wang
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Yunlin Wu
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Jing Ye
- Emergency Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Yiming Lu
- Emergency Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Timothy C Wang
- Department of Medicine, College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA
| | - Chung S Yang
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Shui Ping Tu
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China; Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.
| |
Collapse
|
15
|
Muscari C, Giordano E, Bonafè F, Govoni M, Pasini A, Guarnieri C. Priming adult stem cells by hypoxic pretreatments for applications in regenerative medicine. J Biomed Sci 2013; 20:63. [PMID: 23985033 PMCID: PMC3765890 DOI: 10.1186/1423-0127-20-63] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 08/24/2013] [Indexed: 12/16/2022] Open
Abstract
The efficiency of regenerative medicine can be ameliorated by improving the biological performances of stem cells before their transplantation. Several ex-vivo protocols of non-damaging cell hypoxia have been demonstrated to significantly increase survival, proliferation and post-engraftment differentiation potential of stem cells. The best results for priming cultured stem cells against a following, otherwise lethal, ischemic stress have been obtained with brief intermittent episodes of hypoxia, or anoxia, and reoxygenation in accordance with the extraordinary protection afforded by the conventional maneuver of ischemic preconditioning in severely ischemic organs. These protocols of hypoxic preconditioning can be rather easily reproduced in a laboratory; however, more suitable pharmacological interventions inducing stem cell responses similar to those activated in hypoxia are considered among the most promising solutions for future applications in cell therapy. Here we want to offer an up-to-date review of the molecular mechanisms translating hypoxia into beneficial events for regenerative medicine. To this aim the involvement of epigenetic modifications, microRNAs, and oxidative stress, mainly activated by hypoxia inducible factors, will be discussed. Stem cell adaptation to their natural hypoxic microenvironments (niche) in healthy and neoplastic tissues will be also considered.
Collapse
Affiliation(s)
- Claudio Muscari
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Via Irnerio 48, 40126, Bologna, Italy.
| | | | | | | | | | | |
Collapse
|
16
|
dos Santos RV, da Silva LM. A possible explanation for the variable frequencies of cancer stem cells in tumors. PLoS One 2013; 8:e69131. [PMID: 23950884 PMCID: PMC3737222 DOI: 10.1371/journal.pone.0069131] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Accepted: 06/04/2013] [Indexed: 12/21/2022] Open
Abstract
A controversy surrounds the frequency of cancer stem cells (CSCs) in solid tumors. Initial studies indicated that these cells had a frequency ranging from 0.0001 to 0.1% of the total cells. Recent studies have shown that this does not always seem to be the case. Some of these studies have indicated a frequency of [Formula: see text]. In this paper we propose a stochastic model that is able to capture this potential variability in the frequency of CSCs among the various type of tumors. Considerations regarding the heterogeneity of the tumor cells and its consequences are included. Possible effects on conventional treatments in clinical practice are also described. The model results suggest that traditional attempts to combat cancer cells with rapid cycling can be very stimulating for the cancer stem cell populations.
Collapse
Affiliation(s)
- Renato Vieira dos Santos
- Departamento de Física, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Linaena Méricy da Silva
- Laboratório de Patologia Comparada, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| |
Collapse
|
17
|
Peitzsch C, Kurth I, Kunz-Schughart L, Baumann M, Dubrovska A. Discovery of the cancer stem cell related determinants of radioresistance. Radiother Oncol 2013; 108:378-87. [PMID: 23830195 DOI: 10.1016/j.radonc.2013.06.003] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 05/31/2013] [Accepted: 06/05/2013] [Indexed: 12/23/2022]
Abstract
Tumors are known to be heterogeneous containing a dynamic mixture of phenotypically and functionally different tumor cells. The two concepts attempting to explain the origin of intratumor heterogeneity are the cancer stem cell hypothesis and the clonal evolution model. The stochastic model argues that tumors are biologically homogenous and all cancer cells within the tumor have equal ability to propagate the tumor growth depending on continuing mutations and selective pressure. By contrast, the stem cells model suggests that cancer heterogeneity is due to the hierarchy that originates from a small population of cancer stem cells (CSCs) which are biologically distinct from the bulk tumor and possesses self-renewal, tumorigenic and multilineage potential. Although these two hypotheses have been discussed for a long time as mutually exclusive explanations of tumor heterogeneity, they are easily reconciled serving as a driving force of cancer evolution and diversity. Recent discovery of the cancer cell plasticity and heterogeneity makes the CSC population a moving target that could be hard to track and eradicate. Understanding the signaling mechanisms regulating CSCs during the course of cancer treatment can be indispensable for the optimization of current treatment strategies.
Collapse
Affiliation(s)
- Claudia Peitzsch
- OncoRay National Center for Radiation Research in Oncology, University Hospital/Medical Faculty Carl Gustav Carus, TU Dresden, Germany
| | | | | | | | | |
Collapse
|
18
|
Li Y, Wingert RA. Regenerative medicine for the kidney: stem cell prospects & challenges. Clin Transl Med 2013; 2:11. [PMID: 23688352 PMCID: PMC3665577 DOI: 10.1186/2001-1326-2-11] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 05/14/2013] [Indexed: 12/22/2022] Open
Abstract
The kidney has key roles in maintaining human health. There is an escalating medical crisis in nephrology as growing numbers of patients suffer from kidney diseases that culminate in organ failure. While dialysis and transplantation provide life-saving treatments, these therapies are rife with limitations and place significant burdens on patients and healthcare systems. It has become imperative to find alternative ways to treat existing kidney conditions and preemptive means to stave off renal dysfunction. The creation of innovative medical approaches that utilize stem cells has received growing research attention. In this review, we discuss the regenerative and maladaptive cellular responses that occur during acute and chronic kidney disease, the emerging evidence about renal stem cells, and some of the issues that lie ahead in bridging the gap between basic stem cell biology and regenerative medicine for the kidney.
Collapse
Affiliation(s)
- Yue Li
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rebecca A Wingert
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
19
|
Abstract
The investigation and development of the cancer stem cell (CSC) model has received much focus during these years. CSC is characterized as a small fraction of cancer cells that have an indefinite ability for self-renewal and pluripotency and are responsible for initiating and sustaining of the bulk of cancer. So, whether current treatment strategies, most of which target the rapid division of cancer cells, could interfere with the slow-cycling CSCs is broadly questioned. Meanwhile, however, the new understanding of tumorigenesis has led to the development of new drug screening strategies. Both stem cells and mesenchymal stem cells have been vigorously used in pre-clinical studies of their anti-tumor potential, mainly due to their inherent tumoritropic migratory properties and their ability to carry anti-tumor transgenes. Here, based on the tumorigenic and tumoritropic characteristics of CSCs, we proposed two hypotheses exploring possible usage of CSCs as novel anti-tumor agents and potential sources for tissue regeneration. Further experimental validation of these hypotheses may unravel some new research topics.
Collapse
|
20
|
Cancer stem cell hypothesis: a brief summary and two proposals. Cytotechnology 2012; 65:505-12. [PMID: 23250634 DOI: 10.1007/s10616-012-9517-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 11/16/2012] [Indexed: 12/18/2022] Open
Abstract
The investigation and development of the cancer stem cell (CSC) model has received much focus during these years. CSC is characterized as a small fraction of cancer cells that have an indefinite ability for self-renewal and pluripotency and are responsible for initiating and sustaining of the bulk of cancer. So, whether current treatment strategies, most of which target the rapid division of cancer cells, could interfere with the slow-cycling CSCs is broadly questioned. Meanwhile, however, the new understanding of tumorigenesis has led to the development of new drug screening strategies. Both stem cells and mesenchymal stem cells have been vigorously used in pre-clinical studies of their anti-tumor potential, mainly due to their inherent tumoritropic migratory properties and their ability to carry anti-tumor transgenes. Here, based on the tumorigenic and tumoritropic characteristics of CSCs, we proposed two hypotheses exploring possible usage of CSCs as novel anti-tumor agents and potential sources for tissue regeneration. Further experimental validation of these hypotheses may unravel some new research topics.
Collapse
|
21
|
Yi SY, Hao YB, Nan KJ, Fan TL. Cancer stem cells niche: a target for novel cancer therapeutics. Cancer Treat Rev 2012; 39:290-6. [PMID: 23219150 DOI: 10.1016/j.ctrv.2012.10.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 10/05/2012] [Accepted: 10/08/2012] [Indexed: 12/18/2022]
Abstract
Nowadays, cancer has been a frequent disease, and the first or second most common cause of death worldwide. Despite a better understanding of the biology of cancer cells, the therapy of most cancers has not significantly changed for the past four decades. It is because conventional chemotherapies and/or radiation therapies are usually designed to eradicate highly proliferative cells. Mounting evidence has implicated that cancer is a disease of stem cells. Cancer stem cells (CSC) are often relatively quiescent, and therefore may not be affected by therapies targeting rapidly dividing cells. Like normal stem cells (NSC) residing in a "stem cell niche" that maintains them in a stem-like state, CSC also require a special microenvironment to control their self-renewal and undifferentiated state. The "CSC niche" is likely to be the most crucial target in the treatment of cancer. In this article, we summarize the current knowledge regarding CSC and their niche microenvironments. Understanding of CSC's origin, molecular profile, and interaction with their microenvironments, this could be a paradigm shift in the treatment of cancer, away from targeting the blast cells and towards the targeting of the CSC, thus improving therapeutic outcome.
Collapse
Affiliation(s)
- Shan-Yong Yi
- Department of Oncology of the Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zheng Zhou 450007, Henan Province, China.
| | | | | | | |
Collapse
|
22
|
Abstract
The detailed understanding of adult tissue stem cells has significance for both regenerative medicine and oncology. This perspective will discuss how major advances in our ability to identify and monitor these cells, which include genetic lineage tracing, FACS purification, and robust in vitro clonogenic assays, have changed our view of their roles in many organs. Label retention and quiescence are no longer considered obligatory stem cell features. Furthermore, some tissues have more than one type of stem cell, each used in only particular situations of regenerative stress. Thus, there is no "one size fits all" adult tissue stem cell paradigm.
Collapse
|
23
|
Kunz-Schughart L, Mueller-Klieser W, Vaupel P. Tumorpathophysiologie. Strahlenther Onkol 2012; 188 Suppl 3:299-303. [DOI: 10.1007/s00066-012-0203-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
24
|
Katz SG, Fisher JK, Correll M, Bronson RT, Ligon KL, Walensky LD. Brain and testicular tumors in mice with progenitor cells lacking BAX and BAK. Oncogene 2012; 32:4078-85. [PMID: 22986529 PMCID: PMC3529761 DOI: 10.1038/onc.2012.421] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 07/12/2012] [Accepted: 07/24/2012] [Indexed: 01/08/2023]
Abstract
The pro-apoptotic BCL-2 family proteins BAX and BAK serve as essential gatekeepers of the intrinsic apoptotic pathway and, when activated, transform into pore forming homo-oligomers that permeabilize the mitochondrial outer membrane. Deletion of Bax and Bak causes marked resistance to death stimuli in a variety of cell types. Bax−/−Bak−/− mice are predominantly nonviable and survivors exhibit multiple developmental abnormalities characterized by cellular excess, including accumulation of neural progenitor cells in the periventricular, hippocampal, cerebellar, and olfactory bulb regions of the brain. To explore the long-term pathophysiologic consequences of BAX/BAK deficiency in a stem cell niche, we generated Bak−/− mice with conditional deletion of Bax in Nestin-positive cells. Aged NestinCreBaxfl/flBak−/− mice manifest progressive brain enlargement with a profound accumulation of NeuN- and Sox2-positive neural progenitor cells within the subventricular zone. One-third of the mice develop frank masses comprised of neural progenitors, and in 20% of these cases, more aggressive, hypercellular tumors emerged. Unexpectedly, 60% of NestinCreBaxfl/flBak−/− mice harbored high-grade tumors within the testis, a peripheral site of Nestin expression. This in vivo model of severe apoptotic blockade highlights the constitutive role of BAX/BAK in long-term regulation of Nestin-positive progenitor cell pools, with loss of function predisposing to adult-onset tumorigenesis.
Collapse
Affiliation(s)
- S G Katz
- Department of Pediatric Oncology and the Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | | | | | | | | |
Collapse
|
25
|
Leukemia and benzene. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2012; 9:2875-93. [PMID: 23066403 PMCID: PMC3447593 DOI: 10.3390/ijerph9082875] [Citation(s) in RCA: 160] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 07/05/2012] [Accepted: 08/07/2012] [Indexed: 01/21/2023]
Abstract
Excessive exposure to benzene has been known for more than a century to damage the bone marrow resulting in decreases in the numbers of circulating blood cells, and ultimately, aplastic anemia. Of more recent vintage has been the appreciation that an alternative outcome of benzene exposure has been the development of one or more types of leukemia. While many investigators agree that the array of toxic metabolites, generated in the liver or in the bone marrow, can lead to traumatic bone marrow injury, the more subtle mechanisms leading to leukemia have yet to be critically dissected. This problem appears to have more general interest because of the recognition that so-called "second cancer" that results from prior treatment with alkylating agents to yield tumor remissions, often results in a type of leukemia reminiscent of benzene-induced leukemia. Furthermore, there is a growing literature attempting to characterize the fine structure of the marrow and the identification of so called "niches" that house a variety of stem cells and other types of cells. Some of these "niches" may harbor cells capable of initiating leukemias. The control of stem cell differentiation and proliferation via both inter- and intra-cellular signaling will ultimately determine the fate of these transformed stem cells. The ability of these cells to avoid checkpoints that would prevent them from contributing to the leukemogenic response is an additional area for study. Much of the study of benzene-induced bone marrow damage has concentrated on determining which of the benzene metabolites lead to leukemogenesis. The emphasis now should be directed to understanding how benzene metabolites alter bone marrow cell biology.
Collapse
|
26
|
You N, Liu W, Zhong X, Ji R, Zhang M, You H, Dou K, Tao K. Tg737 inhibition results in malignant transformation in fetal liver stem/progenitor cells by promoting cell-cycle progression and differentiation arrest. Mol Carcinog 2012; 51:659-673. [PMID: 21837759 DOI: 10.1002/mc.20839] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 07/07/2011] [Accepted: 07/11/2011] [Indexed: 12/14/2022]
Abstract
Cancer stem/progenitor cells (CSPCs) may originate from the malignant transformation of normal stem cells. However, the mechanism by which normal stem cells undergo such transformation is not understood. Our previous studies provided evidence that Tg737 may play an important role in carcinogenesis of liver stem cells. In this study, we investigated the role of Tg737 in the malignant transformation of fetal liver stem/progenitor cells (FLSPCs). We inhibited Tg737 in FLSPCs using short hairpin RNA (shRNA). The microscopic observations of freshly purified Tg737 normal FLSPCs (nFLSPCs) and Tg737-silent FLSPCs (sFLSPCs), which showed high expression levels of stem cell markers, revealed no significant morphological changes in sFLSPCs. Following RNAi of Tg737, the mRNA and protein levels of sFLSPCs decreased by 81.81% and 80.10% as shown by PCR, Western blot and immunocytochemistry analyses. Excluding apoptosis-related effects, we found that silencing of Tg737 resulted in enhanced cell proliferation through promoting cell-cycle progression via upregulation of cyclin D1 and cyclin B expression (P < 0.05). Silencing of Tg737 also resulted in significant arrest of cell differentiation (P < 0.05), stable expression of both albumin (ALB) and alpha fetoprotein (AFP) (P > 0.05) and quiescent ultrastructure. Assessment of cell malignant traits by transwell migration assays and by growth of xenograft tumors in athymic mice showed that reduced expression of Tg737 greatly promoted cell invasion and hepatocarcinogenesis of FLSPCs (P < 0.05). This work shows that inactivation of Tg737 may play an important role in malignant transformation of FLSPCs.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Blotting, Western
- Cell Cycle Checkpoints/genetics
- Cell Differentiation/genetics
- Cell Movement/genetics
- Cell Transformation, Neoplastic/genetics
- Cells, Cultured
- Female
- Fetal Stem Cells/metabolism
- Fetal Stem Cells/transplantation
- Fetal Stem Cells/ultrastructure
- Liver/cytology
- Liver/embryology
- Mice
- Mice, Nude
- Microscopy, Electron, Transmission
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- RNA Interference
- Rats
- Rats, Inbred F344
- Reverse Transcriptase Polymerase Chain Reaction
- Transplantation, Heterologous
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
Collapse
Affiliation(s)
- Nan You
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, P. R. China
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Brunner TB, Kunz-Schughart LA, Grosse-Gehling P, Baumann M. Cancer Stem Cells as a Predictive Factor in Radiotherapy. Semin Radiat Oncol 2012; 22:151-74. [DOI: 10.1016/j.semradonc.2011.12.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
28
|
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed and lethal cancers worldwide. It is a multistep process that requires the accumulation of genetic/epigenetic aberrations. There are several issues concerning colorectal carcinogenesis that remain unanswered, such as the cell of origin and the type of cells that propagate the tumor after its initiation. There are two models of carcinogenesis: the stochastic and the cancer stem cell (CSC) model. According to the stochastic model, any kind of cell is capable of initiating and promoting cancer development, whereas the CSC model suggests that tumors are hierarchically organized and only CSCs possess cancer-promoting potential. Moreover, various molecular pathways, such as Wingless/Int (Wnt) and Notch, as well as the complex crosstalk network between microenvironment and CSCs, are involved in CRC. Identification of CSCs remains controversial due to the lack of widely accepted specific molecular markers. CSCs are responsible for tumor relapse, because conventional drugs fail to eliminate the CSC reservoir. Therefore, the design of CSC-targeted interventions is a rational target, which will enhance responsiveness to traditional therapeutic strategies and reduce local recurrence and metastasis. This review discusses the implications of the newly introduced CSC model in CRC, the markers used up to now for CSC identification, and its potential implications in the design of novel therapeutic approaches.
Collapse
|
29
|
Mendrysa SM, Ghassemifar S, Malek R. p53 in the CNS: Perspectives on Development, Stem Cells, and Cancer. Genes Cancer 2011; 2:431-42. [PMID: 21779511 DOI: 10.1177/1947601911409736] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The p53 tumor suppressor potently limits the growth of immature and mature neurons under conditions of cellular stress. Although loss of p53 function contributes to the pathogenesis of central nervous system (CNS) tumors, excessive p53 function is implicated in neural tube defects, embryonic lethality, and neuronal degeneration. Thus, p53 function must be tightly controlled. The anti-proliferative properties of p53 are mediated, in part, through the induction of apoptosis, cell cycle arrest, and senescence. Although there is still much to be learned about the role of p53 in these processes, recent evidence supports exciting new roles for p53 in a wide range of processes, including neural precursor cell self-renewal, differentiation, and cell fate decisions. Understanding the full repertoire of p53 function in CNS development and tumorigenesis may provide us with novel points of therapeutic intervention for human diseases of the CNS.
Collapse
Affiliation(s)
- Susan M Mendrysa
- Department of Basic Medical Sciences, School of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | | | | |
Collapse
|
30
|
Haakensen VD, Lingjaerde OC, Lüders T, Riis M, Prat A, Troester MA, Holmen MM, Frantzen JO, Romundstad L, Navjord D, Bukholm IK, Johannesen TB, Perou CM, Ursin G, Kristensen VN, Børresen-Dale AL, Helland A. Gene expression profiles of breast biopsies from healthy women identify a group with claudin-low features. BMC Med Genomics 2011; 4:77. [PMID: 22044755 PMCID: PMC3216859 DOI: 10.1186/1755-8794-4-77] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 11/01/2011] [Indexed: 12/02/2022] Open
Abstract
Background Increased understanding of the variability in normal breast biology will enable us to identify mechanisms of breast cancer initiation and the origin of different subtypes, and to better predict breast cancer risk. Methods Gene expression patterns in breast biopsies from 79 healthy women referred to breast diagnostic centers in Norway were explored by unsupervised hierarchical clustering and supervised analyses, such as gene set enrichment analysis and gene ontology analysis and comparison with previously published genelists and independent datasets. Results Unsupervised hierarchical clustering identified two separate clusters of normal breast tissue based on gene-expression profiling, regardless of clustering algorithm and gene filtering used. Comparison of the expression profile of the two clusters with several published gene lists describing breast cells revealed that the samples in cluster 1 share characteristics with stromal cells and stem cells, and to a certain degree with mesenchymal cells and myoepithelial cells. The samples in cluster 1 also share many features with the newly identified claudin-low breast cancer intrinsic subtype, which also shows characteristics of stromal and stem cells. More women belonging to cluster 1 have a family history of breast cancer and there is a slight overrepresentation of nulliparous women in cluster 1. Similar findings were seen in a separate dataset consisting of histologically normal tissue from both breasts harboring breast cancer and from mammoplasty reductions. Conclusion This is the first study to explore the variability of gene expression patterns in whole biopsies from normal breasts and identified distinct subtypes of normal breast tissue. Further studies are needed to determine the specific cell contribution to the variation in the biology of normal breasts, how the clusters identified relate to breast cancer risk and their possible link to the origin of the different molecular subtypes of breast cancer.
Collapse
Affiliation(s)
- Vilde D Haakensen
- Dept of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Norway
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Paracrine molecules of mesenchymal stem cells for hematopoietic stem cell niche. BONE MARROW RESEARCH 2011; 2011:353878. [PMID: 22046560 PMCID: PMC3196250 DOI: 10.1155/2011/353878] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 07/26/2011] [Accepted: 07/26/2011] [Indexed: 12/24/2022]
Abstract
Hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs) are both adult stem cells residing in the bone marrow. MSCs interact with HSCs, they stimulate and enhance the proliferation of HSCs by secreting regulatory molecules and cytokines, providing a specialized microenvironment for controlling the process of hematopoiesis. In this paper we discuss how MSCs contribute to HSC niche, maintain the stemness and proliferation of HSCs, and support HSC transplantation.
Collapse
|
32
|
Abstract
Hundreds of G protein coupled receptor (GPCR) isotypes integrate and coordinate the function of individual cells mediating signaling between different organs in our bodies. As an aberration of the normal relationships that organize cells' coexistence, cancer has to deceive cell-cell communication in order to grow and spread. GPCRs play a critical role in this process. Despite the fact that GPCRs represent one of the most common drug targets, current medical practice includes only a few anticancer compounds directly acting on their signaling. Many approaches can be envisaged to target GPCRs involved in oncology. Beyond interfering with GPCRs signaling by using agonists or antagonists to prevent cell proliferation, favor apoptosis, induce maturation, prevent migration, etc., the high specificity of the interaction between the receptors and their ligands can be exploited to deliver toxins, antineoplastic drugs or isotopes to transformed cells. In this review we describe the strategies that are in use, or appear promising, to act directly on GPCRs in the fight against neoplastic transformation and tumor progression.
Collapse
|
33
|
Novel Perspectives on p53 Function in Neural Stem Cells and Brain Tumors. JOURNAL OF ONCOLOGY 2010; 2011:852970. [PMID: 21209724 PMCID: PMC3010739 DOI: 10.1155/2011/852970] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 10/18/2010] [Accepted: 10/29/2010] [Indexed: 12/30/2022]
Abstract
Malignant glioma is the most common brain tumor in adults and is associated with a very poor prognosis. Mutations in the p53 tumor suppressor gene are frequently detected in gliomas. p53 is well-known for its ability to induce cell cycle arrest, apoptosis, senescence, or differentiation following cellular stress. That the guardian of the genome also controls stem cell self-renewal and suppresses pluripotency adds a novel level of complexity to p53. Exactly how p53 works in order to prevent malignant transformation of cells in the central nervous system remains unclear, and despite being one of the most studied proteins, there is a need to acquire further knowledge about p53 in neural stem cells. Importantly, the characterization of glioma cells with stem-like properties, also known as brain tumor stem cells, has opened up for the development of novel targeted therapies. Here, we give an overview of what is currently known about p53 in brain tumors and neural stem cells. Specifically, we review the literature regarding transformation of adult neural stem cells and, we discuss how the loss of p53 and deregulation of growth factor signaling pathways, such as increased PDGF signaling, lead to brain tumor development. Reactivation of p53 in brain tumor stem cell populations in combination with current treatments for glioma should be further explored and may become a viable future therapeutic approach.
Collapse
|
34
|
|