1
|
Majd NK, Vo HH, Moran CA, Weathers SP, Song IW, Williford GL, Rodon J, Fu S, Tsimberidou AM. Metastatic extraneural glioblastoma diagnosed with molecular testing. Oncologist 2024; 29:811-816. [PMID: 38837109 PMCID: PMC11379637 DOI: 10.1093/oncolo/oyae115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 04/19/2024] [Indexed: 06/06/2024] Open
Abstract
Glioblastoma, the most common malignant brain tumor in adults, is associated with a median overall survival duration of less than 2 years. Extraneural metastases occur in less than 1% of all patients with glioblastoma. The mechanism of extraneural metastasis is unclear. We present a case of extensive extraneural, extraosseous, epidural, and soft-tissue metastasis of glioblastoma. The diagnosis of metastatic glioblastoma was made only after next-generation sequencing (NGS) of the metastatic paraspinal lesions was completed. The CDK4, pTERT, PTEN, and TP53 molecular alterations seen in the initial intracranial glioblastoma were found in the paraspinal tumor, along with the addition of MYC, which is implicated in angiogenesis and epidermal-to-mesenchymal transition. Immunohistochemical stains showed that neoplastic cells were negative for GFAP. In conclusion, this case raises awareness about the role of NGS in the diagnosis of extraneural glioblastoma. This diagnosis was not possible with histology alone and only became evident after molecular profiling of the metastatic lesions and its comparison to the original tumor.
Collapse
Affiliation(s)
- Nazanin K Majd
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Henry Hiep Vo
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Cesar A Moran
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Shiao-Pei Weathers
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - I-Wen Song
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Garret L Williford
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jordi Rodon
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Apostolia-Maria Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
2
|
Jiang W, Yang Y, Mercer-Smith AR, Valdivia A, Bago JR, Woodell AS, Buckley AA, Marand MH, Qian L, Anders CK, Hingtgen SD. Development of next-generation tumor-homing induced neural stem cells to enhance treatment of metastatic cancers. SCIENCE ADVANCES 2021; 7:eabf1526. [PMID: 34108203 PMCID: PMC8189583 DOI: 10.1126/sciadv.abf1526] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 04/23/2021] [Indexed: 05/08/2023]
Abstract
Engineered tumor-homing neural stem cells (NSCs) have shown promise in treating cancer. Recently, we transdifferentiated skin fibroblasts into human-induced NSCs (hiNSC) as personalized NSC drug carriers. Here, using a SOX2 and spheroidal culture-based reprogramming strategy, we generated a new hiNSC variant, hiNeuroS, that was genetically distinct from fibroblasts and first-generation hiNSCs and had significantly enhanced tumor-homing and antitumor properties. In vitro, hiNeuroSs demonstrated superior migration to human triple-negative breast cancer (TNBC) cells and in vivo rapidly homed to TNBC tumor foci following intracerebroventricular (ICV) infusion. In TNBC parenchymal metastasis models, ICV infusion of hiNeuroSs secreting the proapoptotic agent TRAIL (hiNeuroS-TRAIL) significantly reduced tumor burden and extended median survival. In models of TNBC leptomeningeal carcinomatosis, ICV dosing of hiNeuroS-TRAIL therapy significantly delayed the onset of tumor formation and extended survival when administered as a prophylactic treatment, as well as reduced tumor volume while prolonging survival when delivered as established tumor therapy.
Collapse
Affiliation(s)
- Wulin Jiang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
| | - Yuchen Yang
- Department of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
| | - Alison R Mercer-Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
| | - Alain Valdivia
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
| | - Juli R Bago
- Department of Hemato-Oncology, University Hospital of Ostrava, 708 52 Ostrava, Czech Republic
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic
| | - Alex S Woodell
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
| | - Andrew A Buckley
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
| | - Michael H Marand
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
| | - Carey K Anders
- Department of Medicine, Duke University, North Carolina, 27710, USA
| | - Shawn D Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA.
- Department of Neurosurgery, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
| |
Collapse
|
3
|
Braun SMG, Petrova R, Tang J, Krokhotin A, Miller EL, Tang Y, Panagiotakos G, Crabtree GR. BAF subunit switching regulates chromatin accessibility to control cell cycle exit in the developing mammalian cortex. Genes Dev 2021; 35:335-353. [PMID: 33602870 PMCID: PMC7919417 DOI: 10.1101/gad.342345.120] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 01/08/2021] [Indexed: 12/28/2022]
Abstract
mSWI/SNF or BAF chromatin regulatory complexes are dosage-sensitive regulators of human neural development frequently mutated in autism spectrum disorders and intellectual disability. Cell cycle exit and differentiation of neural stem/progenitor cells is accompanied by BAF subunit switching to generate neuron-specific nBAF complexes. We manipulated the timing of BAF subunit exchange in vivo and found that early loss of the npBAF subunit BAF53a stalls the cell cycle to disrupt neurogenesis. Loss of BAF53a results in decreased chromatin accessibility at specific neural transcription factor binding sites, including the pioneer factors SOX2 and ASCL1, due to Polycomb accumulation. This results in repression of cell cycle genes, thereby blocking cell cycle progression and differentiation. Cell cycle block upon Baf53a deletion could be rescued by premature expression of the nBAF subunit BAF53b but not by other major drivers of proliferation or differentiation. WNT, EGF, bFGF, SOX2, c-MYC, or PAX6 all fail to maintain proliferation in the absence of BAF53a, highlighting a novel mechanism underlying neural progenitor cell cycle exit in the continued presence of extrinsic proliferative cues.
Collapse
Affiliation(s)
- Simon M G Braun
- Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
- Department of Developmental Biology, Stanford University, California 94305, USA
- Department of Pathology, Stanford University, California 94305, USA
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Ralitsa Petrova
- Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, California 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California 94143, USA
- Kavli Institute for Fundamental Neuroscience, University of California at San Francisco, San Francisco, California 94143, USA
| | - Jiong Tang
- Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
- Department of Developmental Biology, Stanford University, California 94305, USA
- Department of Pathology, Stanford University, California 94305, USA
- Singapore Bioimaging Consortium, Agency for Science Technology and Research (A*STAR), Singapore 138667, Singapore
| | - Andrey Krokhotin
- Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
- Department of Developmental Biology, Stanford University, California 94305, USA
- Department of Pathology, Stanford University, California 94305, USA
| | - Erik L Miller
- Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
- Department of Developmental Biology, Stanford University, California 94305, USA
- Department of Pathology, Stanford University, California 94305, USA
| | - Yitai Tang
- Department of Pathology, Stanford University, California 94305, USA
| | - Georgia Panagiotakos
- Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, California 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California 94143, USA
- Kavli Institute for Fundamental Neuroscience, University of California at San Francisco, San Francisco, California 94143, USA
| | - Gerald R Crabtree
- Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
- Department of Developmental Biology, Stanford University, California 94305, USA
- Department of Pathology, Stanford University, California 94305, USA
| |
Collapse
|
4
|
MYC in Brain Development and Cancer. Int J Mol Sci 2020; 21:ijms21207742. [PMID: 33092025 PMCID: PMC7588885 DOI: 10.3390/ijms21207742] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 12/27/2022] Open
Abstract
The MYC family of transcriptional regulators play significant roles in animal development, including the renewal and maintenance of stem cells. Not surprisingly, given MYC's capacity to promote programs of proliferative cell growth, MYC is frequently upregulated in cancer. Although members of the MYC family are upregulated in nervous system tumours, the mechanisms of how elevated MYC promotes stem cell-driven brain cancers is unknown. If we are to determine how increased MYC might contribute to brain cancer progression, we will require a more complete understanding of MYC's roles during normal brain development. Here, we evaluate evidence for MYC family functions in neural stem cell fate and brain development, with a view to better understand mechanisms of MYC-driven neural malignancies.
Collapse
|
5
|
Pleniceanu O, Harari-Steinberg O, Omer D, Gnatek Y, Lachmi BE, Cohen-Zontag O, Manevitz-Mendelson E, Barzilai A, Yampolsky M, Fuchs Y, Rosenzweig B, Eisner A, Dotan Z, Fine LG, Dekel B, Greenberger S. Successful Introduction of Human Renovascular Units into the Mammalian Kidney. J Am Soc Nephrol 2020; 31:2757-2772. [PMID: 32753400 DOI: 10.1681/asn.2019050508] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 06/22/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Cell-based therapies aimed at replenishing renal parenchyma have been proposed as an approach for treating CKD. However, pathogenic mechanisms involved in CKD such as renal hypoxia result in loss of kidney function and limit engraftment and therapeutic effects of renal epithelial progenitors. Jointly administering vessel-forming cells (human mesenchymal stromal cells [MSCs] and endothelial colony-forming cells [ECFCs]) may potentially result in in vivo formation of vascular networks. METHODS We administered renal tubule-forming cells derived from human adult and fetal kidneys (previously shown to exert a functional effect in CKD mice) into mice, alongside MSCs and ECFCs. We then assessed whether this would result in generation of "renovascular units" comprising both vessels and tubules with potential interaction. RESULTS Directly injecting vessel-forming cells and renal tubule-forming cells into the subcutaneous and subrenal capsular space resulted in self-organization of donor-derived vascular networks that connected to host vasculature, alongside renal tubules comprising tubular epithelia of different nephron segments. Vessels derived from MSCs and ECFCs augmented in vivo tubulogenesis by the renal tubule-forming cells. In vitro coculture experiments showed that MSCs and ECFCs induced self-renewal and genes associated with mesenchymal-epithelial transition in renal tubule-forming cells, indicating paracrine effects. Notably, after renal injury, renal tubule-forming cells and vessel-forming cells infused into the renal artery did not penetrate the renal vascular network to generate vessels; only administering them into the kidney parenchyma resulted in similar generation of human renovascular units in vivo. CONCLUSIONS Combined cell therapy of vessel-forming cells and renal tubule-forming cells aimed at alleviating renal hypoxia and enhancing tubulogenesis holds promise as the basis for new renal regenerative therapies.
Collapse
Affiliation(s)
- Oren Pleniceanu
- The Pediatric Stem Cell Research Institute and Pediatric Nephrology Division, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Orit Harari-Steinberg
- The Pediatric Stem Cell Research Institute and Pediatric Nephrology Division, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Dorit Omer
- The Pediatric Stem Cell Research Institute and Pediatric Nephrology Division, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Yehudit Gnatek
- The Pediatric Stem Cell Research Institute and Pediatric Nephrology Division, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Bat-El Lachmi
- The Pediatric Stem Cell Research Institute and Pediatric Nephrology Division, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Osnat Cohen-Zontag
- The Pediatric Stem Cell Research Institute and Pediatric Nephrology Division, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | | | - Aviv Barzilai
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Dermatology, Sheba Medical Center, Tel Hashomer, Israel
| | - Matan Yampolsky
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Barak Rosenzweig
- Department of Urology, Sheba Medical Center, Tel Hashomer, Israel
| | - Alon Eisner
- Department of Urology, Sheba Medical Center, Tel Hashomer, Israel
| | - Zohar Dotan
- Department of Urology, Sheba Medical Center, Tel Hashomer, Israel
| | - Leon G Fine
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Benjamin Dekel
- The Pediatric Stem Cell Research Institute and Pediatric Nephrology Division, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel .,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shoshana Greenberger
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Dermatology, Sheba Medical Center, Tel Hashomer, Israel
| |
Collapse
|
6
|
Cirotti C, Contadini C, Barilà D. SRC Kinase in Glioblastoma News from an Old Acquaintance. Cancers (Basel) 2020; 12:cancers12061558. [PMID: 32545574 PMCID: PMC7352599 DOI: 10.3390/cancers12061558] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/05/2020] [Accepted: 06/10/2020] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most recalcitrant brain tumors characterized by a tumor microenvironment (TME) that strongly supports GBM growth, aggressiveness, invasiveness, and resistance to therapy. Importantly, a common feature of GBM is the aberrant activation of receptor tyrosine kinases (RTKs) and of their downstream signaling cascade, including the non-receptor tyrosine kinase SRC. SRC is a central downstream intermediate of many RTKs, which triggers the phosphorylation of many substrates, therefore, promoting the regulation of a wide range of different pathways involved in cell survival, adhesion, proliferation, motility, and angiogenesis. In addition to the aforementioned pathways, SRC constitutive activity promotes and sustains inflammation and metabolic reprogramming concurring with TME development, therefore, actively sustaining tumor growth. Here, we aim to provide an updated picture of the molecular pathways that link SRC to these events in GBM. In addition, SRC targeting strategies are discussed in order to highlight strengths and weaknesses of SRC inhibitors in GBM management, focusing our attention on their potentialities in combination with conventional therapeutic approaches (i.e., temozolomide) to ameliorate therapy effectiveness.
Collapse
Affiliation(s)
- Claudia Cirotti
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.C.); (C.C.)
- Laboratory of Signal Transduction, IRCCS-Fondazione Santa Lucia, 00179 Rome, Italy
| | - Claudia Contadini
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.C.); (C.C.)
- Laboratory of Signal Transduction, IRCCS-Fondazione Santa Lucia, 00179 Rome, Italy
| | - Daniela Barilà
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.C.); (C.C.)
- Laboratory of Signal Transduction, IRCCS-Fondazione Santa Lucia, 00179 Rome, Italy
- Correspondence: ; Tel.: +39-065-0170-3168
| |
Collapse
|
7
|
Santoro A, Vlachou T, Luzi L, Melloni G, Mazzarella L, D'Elia E, Aobuli X, Pasi CE, Reavie L, Bonetti P, Punzi S, Casoli L, Sabò A, Moroni MC, Dellino GI, Amati B, Nicassio F, Lanfrancone L, Pelicci PG. p53 Loss in Breast Cancer Leads to Myc Activation, Increased Cell Plasticity, and Expression of a Mitotic Signature with Prognostic Value. Cell Rep 2020; 26:624-638.e8. [PMID: 30650356 PMCID: PMC6334229 DOI: 10.1016/j.celrep.2018.12.071] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/26/2018] [Accepted: 12/14/2018] [Indexed: 12/12/2022] Open
Abstract
Loss of p53 function is invariably associated with cancer. Its role in tumor growth was recently linked to its effects on cancer stem cells (CSCs), although the underlying molecular mechanisms remain unknown. Here, we show that c-myc is a transcriptional target of p53 in mammary stem cells (MaSCs) and is activated in breast tumors as a consequence of p53 loss. Constitutive Myc expression in normal mammary cells leads to increased frequency of MaSC symmetric divisions, extended MaSC replicative-potential, and MaSC-reprogramming of progenitors, whereas Myc activation in breast cancer is necessary and sufficient to maintain the expanding pool of CSCs. Concomitant p53 loss and Myc activation trigger the expression of 189 mitotic genes, which identify patients at high risk of mortality and relapse, independently of other risk factors. Altogether, deregulation of the p53:Myc axis in mammary tumors increases CSC content and plasticity and is a critical determinant of tumor growth and clinical aggressiveness. Myc is overexpressed and deregulated in breast tumors because of p53 signaling attenuation Myc activation favors SC symmetric divisions and SC reprogramming of progenitors Myc activation is necessary and sufficient to sustain the cancer SC phenotype Expression of 189 mitotic p53:Myc targets identifies high-risk breast cancer patients
Collapse
Affiliation(s)
- Angela Santoro
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Thalia Vlachou
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Lucilla Luzi
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Giorgio Melloni
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Via Adamello 16, 20139 Milan, Italy; Department of Biomedical Informatics, Harvard Medical School, 10 Shattuck Street, Boston, MA 02115, USA
| | - Luca Mazzarella
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Errico D'Elia
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Xieraili Aobuli
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Cristina Elisabetta Pasi
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Linsey Reavie
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy; BioPharma Excellence, Agnes-Pockels-Bogen 1, 80922 Munich, Germany
| | - Paola Bonetti
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Via Adamello 16, 20139 Milan, Italy
| | - Simona Punzi
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Lucia Casoli
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Via Adamello 16, 20139 Milan, Italy
| | - Arianna Sabò
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy; Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Via Adamello 16, 20139 Milan, Italy
| | - Maria Cristina Moroni
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Gaetano Ivan Dellino
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20142 Milan, Italy
| | - Bruno Amati
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy; Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Via Adamello 16, 20139 Milan, Italy
| | - Francesco Nicassio
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Via Adamello 16, 20139 Milan, Italy
| | - Luisa Lanfrancone
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Pier Giuseppe Pelicci
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20142 Milan, Italy.
| |
Collapse
|
8
|
Green AL, DeSisto J, Flannery P, Lemma R, Knox A, Lemieux M, Sanford B, O'Rourke R, Ramkissoon S, Jones K, Perry J, Hui X, Moroze E, Balakrishnan I, O'Neill AF, Dunn K, DeRyckere D, Danis E, Safadi A, Gilani A, Hubbell-Engler B, Nuss Z, Levy JMM, Serkova N, Venkataraman S, Graham DK, Foreman N, Ligon K, Jones K, Kung AL, Vibhakar R. BPTF regulates growth of adult and pediatric high-grade glioma through the MYC pathway. Oncogene 2020; 39:2305-2327. [PMID: 31844250 PMCID: PMC7071968 DOI: 10.1038/s41388-019-1125-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 11/15/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023]
Abstract
High-grade gliomas (HGG) afflict both children and adults and respond poorly to current therapies. Epigenetic regulators have a role in gliomagenesis, but a broad, functional investigation of the impact and role of specific epigenetic targets has not been undertaken. Using a two-step, in vitro/in vivo epigenomic shRNA inhibition screen, we determine the chromatin remodeler BPTF to be a key regulator of adult HGG growth. We then demonstrate that BPTF knockdown decreases HGG growth in multiple pediatric HGG models as well. BPTF appears to regulate tumor growth through cell self-renewal maintenance, and BPTF knockdown leads these glial tumors toward more neuronal characteristics. BPTF's impact on growth is mediated through positive effects on expression of MYC and MYC pathway targets. HDAC inhibitors synergize with BPTF knockdown against HGG growth. BPTF inhibition is a promising strategy to combat HGG through epigenetic regulation of the MYC oncogenic pathway.
Collapse
Affiliation(s)
- Adam L Green
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, RC1-N, Mail Stop 8302 12800 E. 19th Ave., Aurora, CO, 80045, USA.
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA.
| | - John DeSisto
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, RC1-N, Mail Stop 8302 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Patrick Flannery
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, RC1-N, Mail Stop 8302 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Rakeb Lemma
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, RC1-N, Mail Stop 8302 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Aaron Knox
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, RC1-N, Mail Stop 8302 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | | | - Bridget Sanford
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, RC1-N, Mail Stop 8302 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Rebecca O'Rourke
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, RC1-N, Mail Stop 8302 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | | | | | | | - Xu Hui
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Erin Moroze
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, RC1-N, Mail Stop 8302 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Ilango Balakrishnan
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, RC1-N, Mail Stop 8302 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | | | | | - Deborah DeRyckere
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta/Emory University, Atlanta, GA, USA
| | - Etienne Danis
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, RC1-N, Mail Stop 8302 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Aaron Safadi
- Department of Radiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ahmed Gilani
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO, USA
| | | | - Zachary Nuss
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, RC1-N, Mail Stop 8302 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Jean M Mulcahy Levy
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, RC1-N, Mail Stop 8302 12800 E. 19th Ave., Aurora, CO, 80045, USA
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA
| | - Natalie Serkova
- Department of Radiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Sujatha Venkataraman
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, RC1-N, Mail Stop 8302 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Douglas K Graham
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta/Emory University, Atlanta, GA, USA
| | - Nicholas Foreman
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, RC1-N, Mail Stop 8302 12800 E. 19th Ave., Aurora, CO, 80045, USA
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA
| | - Keith Ligon
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ken Jones
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, RC1-N, Mail Stop 8302 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Andrew L Kung
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rajeev Vibhakar
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, RC1-N, Mail Stop 8302 12800 E. 19th Ave., Aurora, CO, 80045, USA
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA
| |
Collapse
|
9
|
Neural Transcription Factors in Disease Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:437-462. [PMID: 31900920 DOI: 10.1007/978-3-030-32656-2_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Progression to the malignant state is fundamentally dependent on transcriptional regulation in cancer cells. Optimum abundance of cell cycle proteins, angiogenesis factors, immune evasion markers, etc. is needed for proliferation, metastasis or resistance to treatment. Therefore, dysregulation of transcription factors can compromise the normal prostate transcriptional network and contribute to malignant disease progression.The androgen receptor (AR) is considered to be a key transcription factor in prostate cancer (PCa) development and progression. Consequently, androgen pathway inhibitors (APIs) are currently the mainstay in PCa treatment, especially in castration-resistant prostate cancer (CRPC). However, emerging evidence suggests that with increased administration of potent APIs, prostate cancer can progress to a highly aggressive disease that morphologically resembles small cell carcinoma, which is referred to as neuroendocrine prostate cancer (NEPC), treatment-induced or treatment-emergent small cell prostate cancer. This chapter will review how neuronal transcription factors play a part in inducing a plastic stage in prostate cancer cells that eventually progresses to a more aggressive state such as NEPC.
Collapse
|
10
|
Daekee K, Mi-Jung H, Minjun J, Hee-Jin A, Kwang-Won S, Kyung-Sun K. Generation of Genetically Stable Human Direct-Conversion-Derived Neural Stem Cells Using Quantity Control of Proto-oncogene Expression. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 14:388-397. [PMID: 30731320 PMCID: PMC6365637 DOI: 10.1016/j.omtn.2018.12.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 12/13/2018] [Accepted: 12/13/2018] [Indexed: 12/02/2022]
Abstract
As the human lifespan has increased due to developments in medical technology, the number of patients with neurological diseases has rapidly increased. Therefore, studies on effective treatments for neurological diseases are becoming increasingly important. To perform these studies, it is essential to obtain a large number of patient-derived neural cells. The purpose of the present study was to establish a technology that allows the high-efficiency generation of genetically stable, direct-conversion-derived neural stem cells (dcNSCs) through the expression of a new combination of reprogramming factors, including a proto-oncogene. Specifically, human c-MYC proto-oncogene and the human SOX2 gene were overexpressed in a precisely controlled manner in various human somatic cells. As a result, the direct conversion into multipotent dcNSCs occurred only when the cells were treated with an MOI of 1 of hc-MYC proto-oncogene and hSOX2 retrovirus. When MOIs of 5 or 10 were utilized, distinct results were obtained. In addition, the pluripotency was bypassed during this process. Notably, as the MOI used to treat the cells increased, expression of the p53 tumor suppressor gene, which is typically a reprogramming hurdle, increased proportionately. Interestingly, p53 was genetically stable in dcNSCs generated through direct conversion into a low p53 expression state. In the present study, generation of genetically stable dcNSCs using direct conversion was optimized by precisely controlling the overexpression of a proto-oncogene. This method could be utilized in future studies, such as in vitro drug screening using generated dcNSCs. In addition, this method could be effectively utilized in studies on direct conversion into other types of target cells.
Collapse
Affiliation(s)
- Kwon Daekee
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul 08826, South Korea
| | - Han Mi-Jung
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul 08826, South Korea
| | - Ji Minjun
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul 08826, South Korea
| | - Ahn Hee-Jin
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul 08826, South Korea
| | - Seo Kwang-Won
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul 08826, South Korea
| | - Kang Kyung-Sun
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul 08826, South Korea; Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
11
|
Vinograd-Byk H, Renbaum P, Levy-Lahad E. Vrk1 partial Knockdown in Mice Results in Reduced Brain Weight and Mild Motor Dysfunction, and Indicates Neuronal VRK1 Target Pathways. Sci Rep 2018; 8:11265. [PMID: 30050127 PMCID: PMC6062608 DOI: 10.1038/s41598-018-29215-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/04/2018] [Indexed: 12/12/2022] Open
Abstract
Mutations in Vaccinia-related kinase 1 (VRK1) have emerged as a cause of severe neuronal phenotypes in human, including brain developmental defects and degeneration of spinal motor neurons, leading to Spinal Muscular Atrophy (SMA) or early onset Amyotrophic Lateral Sclerosis (ALS). Vrk1 gene-trap partial Knockout (KO) mice (Vrk1GT3/GT3), which express decreased levels of Vrk1, are sterile due to impaired gamete production. Here, we examined whether this mouse model also presents neuronal phenotypes. We found a 20-50% reduction in Vrk1 expression in neuronal tissues of the Vrk1GT3/GT3 mice, leading to mild neuronal phenotypes including significant but small reduction in brain mass and motor (rotarod) impairment. Analysis of gene expression in the Vrk1GT3/GT3 cortex predicts novel roles for VRK1 in neuronal pathways including neurotrophin signaling, axon guidance and pathways implicated in the pathogenesis of ALS. Together, our studies of the partial KO Vrk1 mice reveal that even moderately reduced levels of Vrk1 expression result in minor neurological impairment and indicate new neuronal pathways likely involving VRK1.
Collapse
Affiliation(s)
- Hadar Vinograd-Byk
- Medical Genetics Institute, Shaare Zedek Medical Center, Jerusalem, 91031, Israel
- Hebrew University Medical School, Jerusalem, 91120, Israel
| | - Paul Renbaum
- Medical Genetics Institute, Shaare Zedek Medical Center, Jerusalem, 91031, Israel
| | - Ephrat Levy-Lahad
- Medical Genetics Institute, Shaare Zedek Medical Center, Jerusalem, 91031, Israel.
- Hebrew University Medical School, Jerusalem, 91120, Israel.
| |
Collapse
|
12
|
Kerosuo L, Bronner ME. cMyc Regulates the Size of the Premigratory Neural Crest Stem Cell Pool. Cell Rep 2017; 17:2648-2659. [PMID: 27926868 DOI: 10.1016/j.celrep.2016.11.025] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 10/03/2016] [Accepted: 11/03/2016] [Indexed: 12/26/2022] Open
Abstract
The neural crest is a transient embryonic population that originates within the central nervous system (CNS) and then migrates into the periphery and differentiates into multiple cell types. The mechanisms that govern neural crest stem-like characteristics and self-renewal ability are poorly understood. Here, we show that the proto-oncogene cMyc is a critical factor in the chick dorsal neural tube, where it regulates the size of the premigratory neural crest stem cell pool. Loss of cMyc dramatically decreases the number of emigrating neural crest cells due to reduced self-renewal capacity, increased cell death, and shorter duration of the emigration process. Interestingly, rather than via E-Box binding, cMyc acts in the dorsal neural tube by interacting with another transcription factor, Miz1, to promote self-renewal. The finding that cMyc operates in a non-canonical manner in the premigratory neural crest highlights the importance of examining its role at specific time points and in an in vivo context.
Collapse
Affiliation(s)
- Laura Kerosuo
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
13
|
Tanno B, Leonardi S, Babini G, Giardullo P, De Stefano I, Pasquali E, Saran A, Mancuso M. Nanog-driven cell-reprogramming and self-renewal maintenance in Ptch1 +/- granule cell precursors after radiation injury. Sci Rep 2017; 7:14238. [PMID: 29079783 PMCID: PMC5660207 DOI: 10.1038/s41598-017-14506-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 10/11/2017] [Indexed: 12/31/2022] Open
Abstract
Medulloblastoma (MB) is the most common pediatric brain tumor, comprising four distinct molecular variants, one of which characterized by activation of the Sonic Hedgehog (SHH) pathway, driving 25–30% of sporadic MB. SHH-dependent MBs arise from granule cell precursors (GCPs), are fatal in 40–70% of cases and radioresistance strongly contributes to poor prognosis and tumor recurrence. Patched1 heterozygous (Ptch1+/−) mice, carrying a germ-line heterozygous inactivating mutation in the Ptch1 gene, the Shh receptor and negative regulator of the pathway, are uniquely susceptible to MB development after radiation damage in neonatal cerebellum. Here, we irradiated ex-vivo GCPs isolated from cerebella of neonatal WT and Ptch1+/− mice. Our results highlight a less differentiated status of Ptch1-mutated cells after irradiation, influencing DNA damage response. Increased expression levels of pluripotency genes Nanog, Oct4 and Sal4, together with greater clonogenic potential, clearly suggest that radiation induces expansion of the stem-like cell compartment through cell-reprogramming and self-renewal maintenance, and that this mechanism is strongly dependent on Nanog. These results contribute to clarify the molecular mechanisms that control radiation-induced Shh-mediated tumorigenesis and may suggest Nanog as a potential target to inhibit for adjuvant radiotherapy in treatment of SHH-dependent MB.
Collapse
Affiliation(s)
- Barbara Tanno
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Rome, Italy
| | - Simona Leonardi
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Rome, Italy
| | | | - Paola Giardullo
- Department of Radiation Physics, Guglielmo Marconi University, Rome, Italy.,Department of Sciences, Roma Tre University, Rome, Italy
| | - Ilaria De Stefano
- Department of Radiation Physics, Guglielmo Marconi University, Rome, Italy
| | - Emanuela Pasquali
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Rome, Italy
| | - Anna Saran
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Rome, Italy.
| | - Mariateresa Mancuso
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Rome, Italy.
| |
Collapse
|
14
|
Piltti KM, Cummings BJ, Carta K, Manughian-Peter A, Worne CL, Singh K, Ong D, Maksymyuk Y, Khine M, Anderson AJ. Live-cell time-lapse imaging and single-cell tracking of in vitro cultured neural stem cells - Tools for analyzing dynamics of cell cycle, migration, and lineage selection. Methods 2017; 133:81-90. [PMID: 29050826 DOI: 10.1016/j.ymeth.2017.10.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 09/26/2017] [Accepted: 10/02/2017] [Indexed: 12/21/2022] Open
Abstract
Neural stem cell (NSC) cultures have been considered technically challenging for time-lapse analysis due to high motility, photosensitivity, and growth at confluent densities. We have tested feasibility of long-term live-cell time-lapse analysis for NSC migration and differentiation studies. Here, we describe a method to study the dynamics of cell cycle, migration, and lineage selection in cultured multipotent mouse or human NSCs using single-cell tracking during a long-term, 7-14 day live-cell time-lapse analysis. We used in-house made PDMS inserts with five microwells on a glass coverslip petri-dish to constrain NSC into the area of acquisition during long-term live-cell imaging. In parallel, we have defined image acquisition settings for single-cell tracking of cell cycle dynamics using Fucci-reporter mouse NSC for 7 days as well as lineage selection and migration using human NSC for 14 days. Overall, we show that adjustments of live-cell analysis settings can extend the time period of single-cell tracking in mouse or human NSC from 24-72 h up to 7-14 days and potentially longer. However, we emphasize that experimental use of repeated fluorescence imaging will require careful consideration of controls during acquisition and analysis.
Collapse
Affiliation(s)
- Katja M Piltti
- Sue & Bill Gross Stem Cell Center, University of California-Irvine, Irvine, CA 92697, USA; Physical & Medical Rehabilitation, University of California-Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments & Neurological Disorders, University of California-Irvine, Irvine, CA 92697, USA.
| | - Brian J Cummings
- Sue & Bill Gross Stem Cell Center, University of California-Irvine, Irvine, CA 92697, USA; Physical & Medical Rehabilitation, University of California-Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments & Neurological Disorders, University of California-Irvine, Irvine, CA 92697, USA; Anatomy & Neurobiology, University of California-Irvine, Irvine, CA 92697, USA
| | - Krystal Carta
- Sue & Bill Gross Stem Cell Center, University of California-Irvine, Irvine, CA 92697, USA
| | - Ayla Manughian-Peter
- Sue & Bill Gross Stem Cell Center, University of California-Irvine, Irvine, CA 92697, USA
| | - Colleen L Worne
- Sue & Bill Gross Stem Cell Center, University of California-Irvine, Irvine, CA 92697, USA
| | - Kulbir Singh
- Sue & Bill Gross Stem Cell Center, University of California-Irvine, Irvine, CA 92697, USA
| | - Danier Ong
- Sue & Bill Gross Stem Cell Center, University of California-Irvine, Irvine, CA 92697, USA
| | - Yuriy Maksymyuk
- Sue & Bill Gross Stem Cell Center, University of California-Irvine, Irvine, CA 92697, USA
| | - Michelle Khine
- Biomedical Engineering, University of California-Irvine, Irvine, CA 92697, USA
| | - Aileen J Anderson
- Sue & Bill Gross Stem Cell Center, University of California-Irvine, Irvine, CA 92697, USA; Physical & Medical Rehabilitation, University of California-Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments & Neurological Disorders, University of California-Irvine, Irvine, CA 92697, USA; Anatomy & Neurobiology, University of California-Irvine, Irvine, CA 92697, USA
| |
Collapse
|
15
|
Cliff TS, Wu T, Boward BR, Yin A, Yin H, Glushka JN, Prestegaard JH, Dalton S. MYC Controls Human Pluripotent Stem Cell Fate Decisions through Regulation of Metabolic Flux. Cell Stem Cell 2017; 21:502-516.e9. [PMID: 28965765 DOI: 10.1016/j.stem.2017.08.018] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 07/27/2017] [Accepted: 08/27/2017] [Indexed: 01/07/2023]
Abstract
As human pluripotent stem cells (hPSCs) exit pluripotency, they are thought to switch from a glycolytic mode of energy generation to one more dependent on oxidative phosphorylation. Here we show that, although metabolic switching occurs during early mesoderm and endoderm differentiation, high glycolytic flux is maintained and, in fact, essential during early ectoderm specification. The elevated glycolysis observed in hPSCs requires elevated MYC/MYCN activity. Metabolic switching during endodermal and mesodermal differentiation coincides with a reduction in MYC/MYCN and can be reversed by ectopically restoring MYC activity. During early ectodermal differentiation, sustained MYCN activity maintains the transcription of "switch" genes that are rate-limiting for metabolic activity and lineage commitment. Our work, therefore, shows that metabolic switching is lineage-specific and not a required step for exit of pluripotency in hPSCs and identifies MYC and MYCN as developmental regulators that couple metabolism to pluripotency and cell fate determination.
Collapse
Affiliation(s)
- Timothy S Cliff
- Department of Biochemistry and Molecular Biology, University of Georgia, 500 D.W. Brooks Drive, Athens, GA 30602, USA; Center for Molecular Medicine, University of Georgia, 500 D.W. Brooks Drive, Athens, GA 30602, USA
| | - Tianming Wu
- Department of Biochemistry and Molecular Biology, University of Georgia, 500 D.W. Brooks Drive, Athens, GA 30602, USA; Center for Molecular Medicine, University of Georgia, 500 D.W. Brooks Drive, Athens, GA 30602, USA
| | - Benjamin R Boward
- Department of Biochemistry and Molecular Biology, University of Georgia, 500 D.W. Brooks Drive, Athens, GA 30602, USA; Center for Molecular Medicine, University of Georgia, 500 D.W. Brooks Drive, Athens, GA 30602, USA
| | - Amelia Yin
- Department of Biochemistry and Molecular Biology, University of Georgia, 500 D.W. Brooks Drive, Athens, GA 30602, USA; Center for Molecular Medicine, University of Georgia, 500 D.W. Brooks Drive, Athens, GA 30602, USA
| | - Hang Yin
- Department of Biochemistry and Molecular Biology, University of Georgia, 500 D.W. Brooks Drive, Athens, GA 30602, USA; Center for Molecular Medicine, University of Georgia, 500 D.W. Brooks Drive, Athens, GA 30602, USA
| | - John N Glushka
- Department of Biochemistry and Molecular Biology, University of Georgia, 500 D.W. Brooks Drive, Athens, GA 30602, USA; Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - James H Prestegaard
- Department of Biochemistry and Molecular Biology, University of Georgia, 500 D.W. Brooks Drive, Athens, GA 30602, USA; Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - Stephen Dalton
- Department of Biochemistry and Molecular Biology, University of Georgia, 500 D.W. Brooks Drive, Athens, GA 30602, USA; Center for Molecular Medicine, University of Georgia, 500 D.W. Brooks Drive, Athens, GA 30602, USA.
| |
Collapse
|
16
|
Kanatsu-Shinohara M, Tanaka T, Ogonuki N, Ogura A, Morimoto H, Cheng PF, Eisenman RN, Trumpp A, Shinohara T. Myc/Mycn-mediated glycolysis enhances mouse spermatogonial stem cell self-renewal. Genes Dev 2017; 30:2637-2648. [PMID: 28007786 PMCID: PMC5204355 DOI: 10.1101/gad.287045.116] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/21/2016] [Indexed: 12/21/2022]
Abstract
Here, Kanatsu-Shinohara et al. investigated the mechanisms underlying Myc regulation of spermatogonial stem cell (SSC) fate. Their findings suggest that Myc-mediated glycolysis is an important factor that increases the frequency of SSC self-renewal division. Myc plays critical roles in the self-renewal division of various stem cell types. In spermatogonial stem cells (SSCs), Myc controls SSC fate decisions because Myc overexpression induces enhanced self-renewal division, while depletion of Max, a Myc-binding partner, leads to meiotic induction. However, the mechanism by which Myc acts on SSC fate is unclear. Here we demonstrate a critical link between Myc/Mycn gene activity and glycolysis in SSC self-renewal. In SSCs, Myc/Mycn are regulated by Foxo1, whose deficiency impairs SSC self-renewal. Myc/Mycn-deficient SSCs not only undergo limited self-renewal division but also display diminished glycolytic activity. While inhibition of glycolysis decreased SSC activity, chemical stimulation of glycolysis or transfection of active Akt1 or Pdpk1 (phosphoinositide-dependent protein kinase 1 ) augmented self-renewal division, and long-term SSC cultures were derived from a nonpermissive strain that showed limited self-renewal division. These results suggested that Myc-mediated glycolysis is an important factor that increases the frequency of SSC self-renewal division.
Collapse
Affiliation(s)
- Mito Kanatsu-Shinohara
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Kyoto 606-8501, Japan
| | - Takashi Tanaka
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | | | - Atsuo Ogura
- Bioresource Center, RIKEN, Tsukuba 305-0074, Japan
| | - Hiroko Morimoto
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Pei Feng Cheng
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Robert N Eisenman
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, Deutsches Krebsforshungszentrum (DKFZ), 69120 Heidelberg, Germany
| | - Takashi Shinohara
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
17
|
Jafari N, Abediankenari S. MicroRNA-34 dysregulation in gastric cancer and gastric cancer stem cell. Tumour Biol 2017; 39:1010428317701652. [PMID: 28468587 DOI: 10.1177/1010428317701652] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer is a major cause of cancer mortality worldwide, with a low survival rate for patients with advanced forms of the disease. Over the recent decades, the investigation of the pathophysiological mechanisms of tumourigenesis has opened promising avenues to understand some of the complexities of cancer treatment. However, tumour regeneration and metastasis impose great difficulty for gastric cancer cure. In recent years, cancer stem cells - a small subset of tumour cells in many cancers - have become a major focus of cancer research. Cancer stem cells are capable of self-renewal and are known to be responsible for tumour initiation, metastasis, therapy resistance and cancer recurrence. Recent studies have revealed the key role of microRNAs - small noncoding RNAs regulating gene expression - in these processes. MicroRNAs play crucial roles in the regulation of a wide range of biological processes in a post-transcriptional manner, though their expression is dysregulated in most malignancies, including gastric cancer. In this article, we review the consequences of aberrant expression of microRNA-34 in cancer and cancer stem cells, with a specific focus on the miR-34 dysregulation in gastric cancer and gastric cancer stem cells. We address the critical effects of the aberrant expression of miR-34 and its target genes in maintaining cancer stem cell properties. Information collection and discussion about the advancements in gastric cancer stem cells and microRNAs can be useful for providing novel insights into patient treatment.
Collapse
Affiliation(s)
- Narjes Jafari
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saeid Abediankenari
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
18
|
Tapia VS, Herrera‐Rojas M, Larrain J. JAK-STAT pathway activation in response to spinal cord injury in regenerative and non-regenerative stages of Xenopus laevis. REGENERATION (OXFORD, ENGLAND) 2017; 4:21-35. [PMID: 28316792 PMCID: PMC5350081 DOI: 10.1002/reg2.74] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/17/2017] [Accepted: 01/18/2017] [Indexed: 12/13/2022]
Abstract
Xenopus laevis tadpoles can regenerate the spinal cord after injury but this capability is lost during metamorphosis. Comparative studies between pre-metamorphic and metamorphic Xenopus stages can aid towards understanding the molecular mechanisms of spinal cord regeneration. Analysis of a previous transcriptome-wide study suggests that, in response to injury, the JAK-STAT pathway is differentially activated in regenerative and non-regenerative stages. We characterized the activation of the JAK-STAT pathway and found that regenerative tadpoles have an early and transient activation. In contrast, the non-regenerative stages have a delayed and sustained activation of the pathway. We found that STAT3 is activated in response to injury mainly in Sox2/3+ ependymal cells, motoneurons and sensory neurons. Finally, to study the role of temporal activation we generated a transgenic line to express a constitutively active version of STAT3. The sustained activation of the JAK-STAT pathway in regenerative tadpoles reduced the expression of pro-neurogenic genes normally upregulated in response to spinal cord injury, suggesting that activation of the JAK-STAT pathway modulates the fate of neural progenitors.
Collapse
Affiliation(s)
- Victor S. Tapia
- Center for Aging and RegenerationMillennium Nucleus in Regenerative BiologyFacultad de Ciencias BiologicasPontificia Universidad Catolica de ChileSantiagoChile
| | - Mauricio Herrera‐Rojas
- Center for Aging and RegenerationMillennium Nucleus in Regenerative BiologyFacultad de Ciencias BiologicasPontificia Universidad Catolica de ChileSantiagoChile
| | - Juan Larrain
- Center for Aging and RegenerationMillennium Nucleus in Regenerative BiologyFacultad de Ciencias BiologicasPontificia Universidad Catolica de ChileSantiagoChile
| |
Collapse
|
19
|
Song J, Oh Y, Kim JY, Cho KJ, Lee JE. Suppression of MicroRNA let-7a Expression by Agmatine Regulates Neural Stem Cell Differentiation. Yonsei Med J 2016; 57:1461-1467. [PMID: 27593875 PMCID: PMC5011279 DOI: 10.3349/ymj.2016.57.6.1461] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 04/27/2016] [Accepted: 04/28/2016] [Indexed: 02/05/2023] Open
Abstract
PURPOSE Neural stem cells (NSCs) effectively reverse some severe central nervous system (CNS) disorders, due to their ability to differentiate into neurons. Agmatine, a biogenic amine, has cellular protective effects and contributes to cellular proliferation and differentiation in the CNS. Recent studies have elucidated the function of microRNA let-7a (let-7a) as a regulator of cell differentiation with roles in regulating genes associated with CNS neurogenesis. MATERIALS AND METHODS This study aimed to investigate whether agmatine modulates the expression of crucial regulators of NSC differentiation including DCX, TLX, c-Myc, and ERK by controlling let-7a expression. RESULTS Our data suggest that high levels of let-7a promoted the expression of TLX and c-Myc, as well as repressed DCX and ERK expression. In addition, agmatine attenuated expression of TLX and increased expression of ERK by negatively regulating let-7a. CONCLUSION Our study therefore enhances the present understanding of the therapeutic potential of NSCs in CNS disorders.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Yumi Oh
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 Plus Project for Medical Sciences and Brain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Youl Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Kyoung Joo Cho
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 Plus Project for Medical Sciences and Brain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 Plus Project for Medical Sciences and Brain Research Institute, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
20
|
Tateishi K, Iafrate AJ, Ho Q, Curry WT, Batchelor TT, Flaherty KT, Onozato ML, Lelic N, Sundaram S, Cahill DP, Chi AS, Wakimoto H. Myc-Driven Glycolysis Is a Therapeutic Target in Glioblastoma. Clin Cancer Res 2016; 22:4452-65. [PMID: 27076630 DOI: 10.1158/1078-0432.ccr-15-2274] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 04/08/2016] [Indexed: 12/21/2022]
Abstract
PURPOSE Deregulated Myc drives an oncogenic metabolic state, including pseudohypoxic glycolysis, adapted for the constitutive production of biomolecular precursors to feed rapid tumor cell growth. In glioblastoma, Myc facilitates renewal of the tumor-initiating cell reservoir contributing to tumor maintenance. We investigated whether targeting the Myc-driven metabolic state could be a selectively toxic therapeutic strategy for glioblastoma. EXPERIMENTAL DESIGN The glycolytic dependency of Myc-driven glioblastoma was tested using (13)C metabolic flux analysis, glucose-limiting culture assays, and glycolysis inhibitors, including inhibitors of the NAD(+) salvage enzyme nicotinamide phosphoribosyl-transferase (NAMPT), in MYC and MYCN shRNA knockdown and lentivirus overexpression systems and in patient-derived glioblastoma tumorspheres with and without MYC/MYCN amplification. The in vivo efficacy of glycolyic inhibition was tested using NAMPT inhibitors in MYCN-amplified patient-derived glioblastoma orthotopic xenograft mouse models. RESULTS Enforced Myc overexpression increased glucose flux and expression of glycolytic enzymes in glioblastoma cells. Myc and N-Myc knockdown and Myc overexpression systems demonstrated that Myc activity determined sensitivity and resistance to inhibition of glycolysis. Small-molecule inhibitors of glycolysis, particularly NAMPT inhibitors, were selectively toxic to MYC/MYCN-amplified patient-derived glioblastoma tumorspheres. NAMPT inhibitors were potently cytotoxic, inducing apoptosis and significantly extended the survival of mice bearing MYCN-amplified patient-derived glioblastoma orthotopic xenografts. CONCLUSIONS Myc activation in glioblastoma generates a dependency on glycolysis and an addiction to metabolites required for glycolysis. Glycolytic inhibition via NAMPT inhibition represents a novel metabolically targeted therapeutic strategy for MYC or MYCN-amplified glioblastoma and potentially other cancers genetically driven by Myc. Clin Cancer Res; 22(17); 4452-65. ©2016 AACR.
Collapse
Affiliation(s)
- Kensuke Tateishi
- Department of Neurosurgery, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - A John Iafrate
- Department of Pathology, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Quan Ho
- Department of Pathology, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - William T Curry
- Department of Neurosurgery, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Tracy T Batchelor
- Division of Hematology/Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts. Stephen E. and Catherine Pappas Center for Neuro-Oncology, Department of Neurology, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Keith T Flaherty
- Division of Hematology/Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Maristela L Onozato
- Department of Pathology, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Nina Lelic
- Department of Neurosurgery, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Sudhandra Sundaram
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Department of Neurology, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Daniel P Cahill
- Department of Neurosurgery, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts.
| | - Andrew S Chi
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York.
| | - Hiroaki Wakimoto
- Department of Neurosurgery, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
21
|
Vo BT, Wolf E, Kawauchi D, Gebhardt A, Rehg JE, Finkelstein D, Walz S, Murphy BL, Youn YH, Han YG, Eilers M, Roussel MF. The Interaction of Myc with Miz1 Defines Medulloblastoma Subgroup Identity. Cancer Cell 2016; 29:5-16. [PMID: 26766587 PMCID: PMC4714043 DOI: 10.1016/j.ccell.2015.12.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 10/25/2015] [Accepted: 12/09/2015] [Indexed: 12/17/2022]
Abstract
Four distinct subgroups of cerebellar medulloblastomas (MBs) differ in their histopathology, molecular profiles, and prognosis. c-Myc (Myc) or MycN overexpression in granule neuron progenitors (GNPs) induces Group 3 (G3) or Sonic Hedgehog (SHH) MBs, respectively. Differences in Myc and MycN transcriptional profiles depend, in part, on their interaction with Miz1, which binds strongly to Myc but not MycN, to target sites on chromatin. Myc suppresses ciliogenesis and reprograms the transcriptome of SHH-dependent GNPs through Miz1-dependent gene repression to maintain stemness. Genetic disruption of the Myc/Miz1 interaction inhibited G3 MB development. Target genes of Myc/Miz1 are repressed in human G3 MBs but not in other subgroups. Therefore, the Myc/Miz1 interaction is a defining hallmark of G3 MB development.
Collapse
Affiliation(s)
- BaoHan T. Vo
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Elmar Wolf
- Theodor Boveri Institute, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Daisuke Kawauchi
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, 262 Danny Thomas Place, Memphis, TN 38105, USA
- German Cancer Research Center (DKFZ), Division of Pediatric Neurooncology (B062) Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Anneli Gebhardt
- Theodor Boveri Institute, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Jerold E. Rehg
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Susanne Walz
- Theodor Boveri Institute, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Josef-Schneider-Str.6, 97080 Würzburg, Germany
| | - Brian L. Murphy
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Yong Ha Youn
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Young-Goo Han
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Martin Eilers
- Theodor Boveri Institute, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Josef-Schneider-Str.6, 97080 Würzburg, Germany
- Correspondence: (M.F.R.); (M.E.)
| | - Martine F. Roussel
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, 262 Danny Thomas Place, Memphis, TN 38105, USA
- Correspondence: (M.F.R.); (M.E.)
| |
Collapse
|
22
|
Abstract
Myc and its paralog MycN are thought to be functionally redundant, but Myc- and MycN-driven medulloblastomas exhibit distinct phenotypes. In this issue of Cancer Cell, Vo and colleagues (2016) show that this phenotypic difference stems from the preferential ability of Myc, relative to MycN, to bind Miz1 and repress transcription.
Collapse
Affiliation(s)
- Haritha Mathsyaraja
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Robert N Eisenman
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| |
Collapse
|
23
|
Winiecka-Klimek M, Smolarz M, Walczak MP, Zieba J, Hulas-Bigoszewska K, Kmieciak B, Piaskowski S, Rieske P, Grzela DP, Stoczynska-Fidelus E. SOX2 and SOX2-MYC Reprogramming Process of Fibroblasts to the Neural Stem Cells Compromised by Senescence. PLoS One 2015; 10:e0141688. [PMID: 26535892 PMCID: PMC4633175 DOI: 10.1371/journal.pone.0141688] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 10/11/2015] [Indexed: 11/19/2022] Open
Abstract
Tumorigenic potential of induced pluripotent stem cells (iPSCs) infiltrating population of induced neural stem cells (iNSCs) generated from iPSCs may limit their medical applications. To overcome such a difficulty, direct reprogramming of adult somatic cells into iNSCs was proposed. The aim of this study was the systematic comparison of induced neural cells (iNc) obtained with different methods—direct reprogramming of human adult fibroblasts with either SOX2 (SiNSc-like) or SOX2 and c-MYC (SMiNSc-like) and induced pluripotent stem cells differentiation to ebiNSc—in terms of gene expression profile, differentiation potential as well as proliferation properties. Immunocytochemistry and real-time PCR analyses were used to evaluate gene expression profile and differentiation potential of various iNc types. Bromodeoxyuridine (BrdU) incorporation and senescence-associated beta-galactosidase (SA-β-gal) assays were used to estimate proliferation potential. All three types of iNc were capable of neuronal differentiation; however, astrocytic differentiation was possible only in case of ebiNSc. Contrary to ebiNSc generation, the direct reprogramming was rarely a propitious process, despite 100% transduction efficiency. The potency of direct iNSCs-like cells generation was lower as compared to iNSCs obtained by iPSCs differentiation, and only slightly improved when c-MYC was added. Directly reprogrammed iNSCs-like cells were lacking the ability to differentiate into astrocytic cells and characterized by poor efficiency of neuronal cells formation. Such features indicated that these cells could not be fully reprogrammed, as confirmed mainly with senescence detection. Importantly, SiNSc-like and SMiNSc-like cells were unable to achieve the long-term survival and became senescent, which limits their possible therapeutic applicability. Our results suggest that iNSCs-like cells, generated in the direct reprogramming attempts, were either not fully reprogrammed or reprogrammed only into neuronal progenitors, mainly because of the inaccuracies of currently available protocols.
Collapse
Affiliation(s)
- Marta Winiecka-Klimek
- Department of Research and Development, Celther Polska, Lodz, Poland
- Department of Tumor Biology, Medical University of Lodz, Lodz, Poland
- * E-mail:
| | - Maciej Smolarz
- Department of Research and Development, Celther Polska, Lodz, Poland
- Department of Tumor Biology, Medical University of Lodz, Lodz, Poland
| | - Maciej P. Walczak
- Department of Research and Development, Celther Polska, Lodz, Poland
| | - Jolanta Zieba
- Department of Research and Development, Celther Polska, Lodz, Poland
- Department of Tumor Biology, Medical University of Lodz, Lodz, Poland
| | | | - Blazej Kmieciak
- Department of Medical Law, Chair of Human Sciences, Medical University of Lodz, Lodz, Poland
| | - Sylwester Piaskowski
- Department of Research and Development, Celther Polska, Lodz, Poland
- Department of Tumor Biology, Medical University of Lodz, Lodz, Poland
| | - Piotr Rieske
- Department of Research and Development, Celther Polska, Lodz, Poland
- Department of Tumor Biology, Medical University of Lodz, Lodz, Poland
| | - Dawid P. Grzela
- Department of Research and Development, Celther Polska, Lodz, Poland
| | - Ewelina Stoczynska-Fidelus
- Department of Research and Development, Celther Polska, Lodz, Poland
- Department of Tumor Biology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
24
|
Xia B, Zhang L, Guo SQ, Li XW, Qu FL, Zhao HF, Zhang LY, Sun BC, You J, Zhang YZ. Coexpression of MYC and BCL-2 predicts prognosis in primary gastrointestinal diffuse large B-cell lymphoma. World J Gastroenterol 2015; 21:2433-2442. [PMID: 25741152 PMCID: PMC4342921 DOI: 10.3748/wjg.v21.i8.2433] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 11/06/2014] [Accepted: 12/22/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether MYC and BCL-2 coexpression has prognostic significance in primary gastrointestinal diffuse large B-cell lymphoma (PGI-DLBCL) patients, and explore its associations with patients’ clinical parameters.
METHODS: Fresh and paraffin-embedded tumor tissue samples from 60 PGI-DLBCL patients who had undergone surgery at the Tianjin Medical University Cancer Institute and Hospital from January 2005 to May 2010 were obtained, and 30 lymphoid tissue samples from reactive lymph nodes of age- and sex-matched patients represented control samples. Staging and diagnostic procedures were conducted according to the Lugano staging system. All patients had been treated with three therapeutic modalities: surgery, chemotherapy, or radiotherapy. Expression of MYC and BCL-2 were detected at both protein and mRNA levels by immunohistochemistry and real-time RT-PCR.
RESULTS: Positive expression levels of MYC and BCL-2 proteins were detected in 35% and 45% of patients, respectively. MYC+/BCL-2+ protein was present in 30% of patients. MYC and BCL-2 protein levels were correlated with high MYC and BCL-2 mRNA expression, respectively (both P < 0.05). We found that advanced-stage disease (at IIE-IV) was associated with MYC and BCL-2 coexpression levels (P < 0.05). In addition, MYC+/BCL-2+ patients had more difficulty in achieving complete remission than others (P < 0.05). Presence of MYC protein expression only affected overall survival and progression-free survival (PFS) when BCL-2 protein was coexpressed. The adverse prognostic impact of MYC+/BCL-2+ protein on PFS remained significant (P < 0.05) even after adjusting for age, Lugano stage, international prognostic index, and BCL-2 protein expression in a multivariable model.
CONCLUSION: MYC+/BCL-2+ patients have worse chemotherapy response and poorer prognosis than patients who only express one of the two proteins, suggesting that assessment of MYC and BCL-2 expression by immunohistochemistry has clinical significance in predicting clinical outcomes of PGI-DLBCL patients.
Collapse
MESH Headings
- Adult
- Aged
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Case-Control Studies
- Chi-Square Distribution
- China
- Disease-Free Survival
- Female
- Gastrointestinal Neoplasms/chemistry
- Gastrointestinal Neoplasms/genetics
- Gastrointestinal Neoplasms/mortality
- Gastrointestinal Neoplasms/pathology
- Gastrointestinal Neoplasms/therapy
- Hospitals, University
- Humans
- Immunohistochemistry
- Kaplan-Meier Estimate
- Lymphoma, Large B-Cell, Diffuse/chemistry
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/therapy
- Male
- Middle Aged
- Multivariate Analysis
- Neoplasm Staging
- Predictive Value of Tests
- Proportional Hazards Models
- Proto-Oncogene Proteins c-bcl-2/analysis
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-myc/analysis
- Proto-Oncogene Proteins c-myc/genetics
- RNA, Messenger/analysis
- Real-Time Polymerase Chain Reaction
- Risk Factors
- Time Factors
- Treatment Outcome
- Young Adult
Collapse
|
25
|
Sanz-Moreno A, Fuhrmann D, Zankel A, Reingruber H, Kern L, Meijer D, Niemann A, Elsässer HP. Late onset neuropathy with spontaneous clinical remission in mice lacking the POZ domain of the transcription factor Myc-interacting zinc finger protein 1 (Miz1) in Schwann cells. J Biol Chem 2014; 290:727-43. [PMID: 25416780 DOI: 10.1074/jbc.m114.605931] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The transcription factor Miz1 (Myc-interacting zinc finger 1) is a known regulator of the cell cycle but also has cell cycle-independent functions. Here we analyzed the role of Miz1 in the peripheral nervous system, using an early embryonic conditional knock-out model in which the Miz1 POZ domain is ablated in Schwann cells. Although the development of myelinated nerve fibers was not impaired, Miz1ΔPOZ mice acquired behavioral signs of a peripheral neuropathy at the age of 3 months. At this time, ultrastructural analysis of the sciatic nerve showed de- and dysmyelination of fibers, with massive outfoldings and a focal infiltration of macrophages. Although the expression of genes encoding structural myelin proteins, such as periaxin, myelin basic protein, and myelin protein zero, was decreased, genes associated with a negative regulation of myelination, including c-Jun, Sox2, and Id2, were up-regulated in Miz1ΔPOZ mice compared with controls. In animals older than 4 months, the motor disabilities vanished, and the ultrastructure of the sciatic nerve exhibited numerous tomacula and remyelinated fibers, as indicated by thinner myelin. No second acute attack was observed up to the age of 1 year. Thus, the deletion of the Miz1 POZ domain in Schwann cells induces an acute neuropathy with a subsequent regeneration in which there is ongoing balancing between de- and remyelination. Miz1ΔPOZ mice are impaired in the maintenance of myelinated fibers and are a promising model for studying remyelination in adult peripheral nerves.
Collapse
Affiliation(s)
- Adrián Sanz-Moreno
- From the Department of Cytobiology and Cytopathobiology, Philipps University of Marburg, Robert-Koch-Strasse 6, 35033 Marburg, Germany
| | - David Fuhrmann
- From the Department of Cytobiology and Cytopathobiology, Philipps University of Marburg, Robert-Koch-Strasse 6, 35033 Marburg, Germany
| | - Armin Zankel
- Graz University of Technology, 8010 Graz, Austria
| | | | - Lara Kern
- From the Department of Cytobiology and Cytopathobiology, Philipps University of Marburg, Robert-Koch-Strasse 6, 35033 Marburg, Germany
| | - Dies Meijer
- Erasmus Medical Center, 3015GE Rotterdam, Netherlands, and
| | | | - Hans-Peter Elsässer
- From the Department of Cytobiology and Cytopathobiology, Philipps University of Marburg, Robert-Koch-Strasse 6, 35033 Marburg, Germany,
| |
Collapse
|
26
|
Annibali D, Whitfield JR, Favuzzi E, Jauset T, Serrano E, Cuartas I, Redondo-Campos S, Folch G, Gonzàlez-Juncà A, Sodir NM, Massó-Vallés D, Beaulieu ME, Swigart LB, Mc Gee MM, Somma MP, Nasi S, Seoane J, Evan GI, Soucek L. Myc inhibition is effective against glioma and reveals a role for Myc in proficient mitosis. Nat Commun 2014; 5:4632. [PMID: 25130259 PMCID: PMC4143920 DOI: 10.1038/ncomms5632] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 07/09/2014] [Indexed: 12/21/2022] Open
Abstract
Gliomas are the most common primary tumours affecting the adult central nervous system and respond poorly to standard therapy. Myc is causally implicated in most human tumours and the majority of glioblastomas have elevated Myc levels. Using the Myc dominant negative Omomyc, we previously showed that Myc inhibition is a promising strategy for cancer therapy. Here, we preclinically validate Myc inhibition as a therapeutic strategy in mouse and human glioma, using a mouse model of spontaneous multifocal invasive astrocytoma and its derived neuroprogenitors, human glioblastoma cell lines, and patient-derived tumours both in vitro and in orthotopic xenografts. Across all these experimental models we find that Myc inhibition reduces proliferation, increases apoptosis and remarkably, elicits the formation of multinucleated cells that then arrest or die by mitotic catastrophe, revealing a new role for Myc in the proficient division of glioma cells. Myc has been implicated in the development of multiple types of cancer. Here, the authors explore the therapeutic potential and mechanism of action of Myc inhibition in mouse and human models of glioblastoma, an aggressive type of tumour that is often resistant to conventional therapy.
Collapse
Affiliation(s)
- Daniela Annibali
- 1] Department of Pathology, Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94143, USA [2] Istituto di Biologia, Medicina Molecolare e NanoBiotecnologie, C.N.R., Dipartimento di Biologia e Biotecnologie, Università La Sapienza, 00185 Rome, Italy [3]
| | - Jonathan R Whitfield
- 1] Vall d'Hebron Institute of Oncology (VHIO), Edifici Mediterrània, Hospital Vall d'Hebron, 08035 Barcelona, Spain [2] Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), 08193 Barcelona, Spain [3]
| | - Emilia Favuzzi
- Istituto di Biologia, Medicina Molecolare e NanoBiotecnologie, C.N.R., Dipartimento di Biologia e Biotecnologie, Università La Sapienza, 00185 Rome, Italy
| | - Toni Jauset
- 1] Vall d'Hebron Institute of Oncology (VHIO), Edifici Mediterrània, Hospital Vall d'Hebron, 08035 Barcelona, Spain [2] Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), 08193 Barcelona, Spain
| | - Erika Serrano
- 1] Vall d'Hebron Institute of Oncology (VHIO), Edifici Mediterrània, Hospital Vall d'Hebron, 08035 Barcelona, Spain [2] Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), 08193 Barcelona, Spain
| | - Isabel Cuartas
- 1] Vall d'Hebron Institute of Oncology (VHIO), Edifici Mediterrània, Hospital Vall d'Hebron, 08035 Barcelona, Spain [2] Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), 08193 Barcelona, Spain
| | - Sara Redondo-Campos
- 1] Vall d'Hebron Institute of Oncology (VHIO), Edifici Mediterrània, Hospital Vall d'Hebron, 08035 Barcelona, Spain [2] Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), 08193 Barcelona, Spain
| | - Gerard Folch
- 1] Vall d'Hebron Institute of Oncology (VHIO), Edifici Mediterrània, Hospital Vall d'Hebron, 08035 Barcelona, Spain [2] Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), 08193 Barcelona, Spain
| | - Alba Gonzàlez-Juncà
- 1] Vall d'Hebron Institute of Oncology (VHIO), Edifici Mediterrània, Hospital Vall d'Hebron, 08035 Barcelona, Spain [2] Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), 08193 Barcelona, Spain
| | - Nicole M Sodir
- 1] Department of Pathology, Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94143, USA [2] Department of Biochemistry, Sanger Building, University of Cambridge, Cambridge CB2 1QW, UK
| | - Daniel Massó-Vallés
- 1] Vall d'Hebron Institute of Oncology (VHIO), Edifici Mediterrània, Hospital Vall d'Hebron, 08035 Barcelona, Spain [2] Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), 08193 Barcelona, Spain
| | - Marie-Eve Beaulieu
- 1] Vall d'Hebron Institute of Oncology (VHIO), Edifici Mediterrània, Hospital Vall d'Hebron, 08035 Barcelona, Spain [2] Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), 08193 Barcelona, Spain
| | - Lamorna B Swigart
- Department of Pathology, Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94143, USA
| | - Margaret M Mc Gee
- UCD School of Biomolecular &Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Maria Patrizia Somma
- Istituto di Biologia, Medicina Molecolare e NanoBiotecnologie, C.N.R., Dipartimento di Biologia e Biotecnologie, Università La Sapienza, 00185 Rome, Italy
| | - Sergio Nasi
- Istituto di Biologia, Medicina Molecolare e NanoBiotecnologie, C.N.R., Dipartimento di Biologia e Biotecnologie, Università La Sapienza, 00185 Rome, Italy
| | - Joan Seoane
- 1] Vall d'Hebron Institute of Oncology (VHIO), Edifici Mediterrània, Hospital Vall d'Hebron, 08035 Barcelona, Spain [2] Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), 08193 Barcelona, Spain [3] Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Gerard I Evan
- Department of Biochemistry, Sanger Building, University of Cambridge, Cambridge CB2 1QW, UK
| | - Laura Soucek
- 1] Vall d'Hebron Institute of Oncology (VHIO), Edifici Mediterrània, Hospital Vall d'Hebron, 08035 Barcelona, Spain [2] Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), 08193 Barcelona, Spain
| |
Collapse
|
27
|
Kim DY, Rhee I, Paik J. Metabolic circuits in neural stem cells. Cell Mol Life Sci 2014; 71:4221-41. [PMID: 25037158 DOI: 10.1007/s00018-014-1686-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 06/25/2014] [Accepted: 07/14/2014] [Indexed: 12/15/2022]
Abstract
Metabolic activity indicative of cellular demand is emerging as a key player in cell fate decision. Numerous studies have demonstrated that diverse metabolic pathways have a critical role in the control of the proliferation, differentiation and quiescence of stem cells. The identification of neural stem/progenitor cells (NSPCs) and the characterization of their development and fate decision process have provided insight into the regenerative potential of the adult brain. As a result, the potential of NSPCs in cell replacement therapies for neurological diseases is rapidly growing. The aim of this review is to discuss the recent findings on the crosstalk among key regulators of NSPC development and the metabolic regulation crucial for the function and cell fate decisions of NSPCs. Fundamental understanding of the metabolic circuits in NSPCs may help to provide novel approaches for reactivating neurogenesis to treat degenerative brain conditions and cognitive decline.
Collapse
Affiliation(s)
- Do-Yeon Kim
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York City, NY, 10065, USA
| | | | | |
Collapse
|
28
|
Lubanska D, Porter LA. The atypical cell cycle regulator Spy1 suppresses differentiation of the neuroblastoma stem cell population. Oncoscience 2014; 1:336-48. [PMID: 25594028 PMCID: PMC4278303 DOI: 10.18632/oncoscience.36] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 05/04/2014] [Indexed: 12/28/2022] Open
Abstract
Neuroblastoma is an aggressive pediatric cancer originating embryonically from the neural crest. The heterogeneity of the disease, as most solid tumors, complicates diagnosis and treatment. In neuroblastoma this heterogeneity is well represented in both primary tumours and derived cell lines and has been shown to be driven by a population of stem-like tumour initiating cells. Resolving the molecular mediators driving the division of this population of cells may indicate effective therapeutic options for neuroblastoma patients. This study has determined that the atypical cyclin-like protein Spy1, recently indicated in driving symmetric division of glioma stem cells, is a critical factor in the stem-like properties of neuroblastoma tumor initiating cell populations. Spy1 activates Cyclin Dependent Kinases (CDK) in a manner that is unique from classical cyclins. Hence this discovery may represent an important opportunity to design CDK inhibitor drugs to uniquely target subpopulations of cells within these aggressive neural tumours.
Collapse
Affiliation(s)
- Dorota Lubanska
- Department of Biological Sciences University of Windsor OntarioWindsor, ON
| | - Lisa A. Porter
- Department of Biological Sciences University of Windsor OntarioWindsor, ON
| |
Collapse
|
29
|
Levinson M, Zhou Q. A penalized Bayesian approach to predicting sparse protein-DNA binding landscapes. ACTA ACUST UNITED AC 2014; 30:636-43. [PMID: 24115169 DOI: 10.1093/bioinformatics/btt585] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
MOTIVATION Cellular processes are controlled, directly or indirectly, by the binding of hundreds of different DNA binding factors (DBFs) to the genome. One key to deeper understanding of the cell is discovering where, when and how strongly these DBFs bind to the DNA sequence. Direct measurement of DBF binding sites (BSs; e.g. through ChIP-Chip or ChIP-Seq experiments) is expensive, noisy and not available for every DBF in every cell type. Naive and most existing computational approaches to detecting which DBFs bind in a set of genomic regions of interest often perform poorly, due to the high false discovery rates and restrictive requirements for prior knowledge. RESULTS We develop SparScape, a penalized Bayesian method for identifying DBFs active in the considered regions and predicting a joint probabilistic binding landscape. Using a sparsity-inducing penalization, SparScape is able to select a small subset of DBFs with enriched BSs in a set of DNA sequences from a much larger candidate set. This substantially reduces the false positives in prediction of BSs. Analysis of ChIP-Seq data in mouse embryonic stem cells and simulated data show that SparScape dramatically outperforms the naive motif scanning method and the comparable computational approaches in terms of DBF identification and BS prediction. AVAILABILITY AND IMPLEMENTATION SparScape is implemented in C++ with OpenMP (optional at compilation) and is freely available at 'www.stat.ucla.edu/∼zhou/Software.html' for academic use.
Collapse
Affiliation(s)
- Matthew Levinson
- Department of Statistics, University of California, Los Angeles, CA 90095, USA
| | | |
Collapse
|
30
|
Abstract
MYC dimerizes with MAX to bind DNA, with a preference for the E-box consensus CACGTG and several variant motifs. In cells, MYC binds DNA preferentially within transcriptionally active promoter regions. Although several thousand promoters are bound under physiological (low MYC) conditions, these represent only a fraction of all accessible, active promoters. MYC overexpression-as commonly observed in cancer cells-leads to invasion of virtually all active promoters, as well as of distal enhancer elements. We summarize here what is currently known about the mechanisms that may guide this process. We propose that binding site recognition is determined by low-affinity protein-protein interactions between MYC/MAX dimers and components of the basal transcriptional machinery, other chromatin-associated protein complexes, and/or DNA-bound transcription factors. DNA binding occurs subsequently, without an obligate requirement for sequence recognition. Local DNA scanning then leads to preferential stabilization of the MYC/MAX dimer on high-affinity DNA elements. This model is consistent with the invasion of all active promoters that occurs at elevated MYC levels, but posits that important differences in affinity persist between physiological target sites and the newly invaded elements, which may not all be bound in a productive regulatory mode. The implications of this model for transcriptional control by MYC in normal and cancer cells are discussed in the light of the latest literature.
Collapse
Affiliation(s)
- Arianna Sabò
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia, 20139 Milan, Italy
| | | |
Collapse
|
31
|
Lubanska D, Market-Velker BA, deCarvalho AC, Mikkelsen T, Fidalgo da Silva E, Porter LA. The cyclin-like protein Spy1 regulates growth and division characteristics of the CD133+ population in human glioma. Cancer Cell 2014; 25:64-76. [PMID: 24434210 DOI: 10.1016/j.ccr.2013.12.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Revised: 05/17/2013] [Accepted: 12/13/2013] [Indexed: 11/17/2022]
Abstract
The heterogeneity of brain cancers, as most solid tumors, complicates diagnosis and treatment. Identifying and targeting populations of cells driving tumorigenesis is a top priority for the cancer biology field. This is not a trivial task; considerable variance exists in the driving mutations, identifying markers, and evolutionary pressures influencing initiating cells in different individual tumors. Despite this, the ability to self-renew and differentiate must be conserved to reseed a heterogeneous tumor mass. Focusing on one example of a tumor-initiating cell population, we demonstrate that the atypical cyclin-like protein Spy1 plays a role in balancing the division properties of glioma cells with stemness properties. This mechanistic insight may provide new opportunities for therapeutic intervention of brain cancer.
Collapse
Affiliation(s)
- Dorota Lubanska
- Department of Biological Sciences, University of Windsor Ontario, Windsor, ON N9B 3P4, Canada
| | - Brenna A Market-Velker
- Department of Biological Sciences, University of Windsor Ontario, Windsor, ON N9B 3P4, Canada
| | - Ana C deCarvalho
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Tom Mikkelsen
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI 48202, USA
| | | | - Lisa A Porter
- Department of Biological Sciences, University of Windsor Ontario, Windsor, ON N9B 3P4, Canada.
| |
Collapse
|
32
|
Kerosuo L, Bronner ME. Biphasic influence of Miz1 on neural crest development by regulating cell survival and apical adhesion complex formation in the developing neural tube. Mol Biol Cell 2013; 25:347-55. [PMID: 24307680 PMCID: PMC3907275 DOI: 10.1091/mbc.e13-06-0327] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Myc interacting zinc finger protein-1 (Miz1) is a transcription factor known to regulate cell cycle- and cell adhesion-related genes in cancer. Here we show that Miz1 also plays a critical role in neural crest development. In the chick, Miz1 is expressed throughout the neural plate and closing neural tube. Its morpholino-mediated knockdown affects neural crest precursor survival, leading to reduction of neural plate border and neural crest specifier genes Msx-1, Pax7, FoxD3, and Sox10. Of interest, Miz1 loss also causes marked reduction of adhesion molecules (N-cadherin, cadherin6B, and α1-catenin) with a concomitant increase of E-cadherin in the neural folds, likely leading to delayed and decreased neural crest emigration. Conversely, Miz1 overexpression results in up-regulation of cadherin6B and FoxD3 expression in the neural folds/neural tube, leading to premature neural crest emigration and increased number of migratory crest cells. Although Miz1 loss effects cell survival and proliferation throughout the neural plate, the neural progenitor marker Sox2 was unaffected, suggesting a neural crest-selective effect. The results suggest that Miz1 is important not only for survival of neural crest precursors, but also for maintenance of integrity of the neural folds and tube, via correct formation of the apical adhesion complex therein.
Collapse
Affiliation(s)
- Laura Kerosuo
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | | |
Collapse
|
33
|
Ilieva M, Dufva M. SOX2 and OCT4 mRNA-expressing cells, detected by molecular beacons, localize to the center of neurospheres during differentiation. PLoS One 2013; 8:e73669. [PMID: 24013403 PMCID: PMC3754928 DOI: 10.1371/journal.pone.0073669] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 07/19/2013] [Indexed: 12/26/2022] Open
Abstract
Neurospheres are used as in vitro assay to measure the properties of neural stem cells. To investigate the molecular and phenotypic heterogeneity of neurospheres, molecular beacons (MBs) targeted against the stem cell markers OCT4 and SOX2 were designed, and synthesized with a 2'-O-methyl RNA backbone. OCT4 and SOX2 MBs were transfected into human embryonic mesencephalon derived cells, which spontaneously form neurospheres when grown on poly-L-ornitine/fibronectin matrix and medium complemented with bFGF. OCT4 and SOX2 gene expression were tracked in individual cell using the MBs. Quantitative image analysis every day for seven days showed that the OCT4 and SOX2 mRNA-expressing cells clustered in the centre of the neurospheres cultured in differentiation medium. By contrast, cells at the periphery of the differentiating spheres developed neurite outgrowths and expressed the tyrosine hydroxylase protein, indicating terminal differentiation. Neurospheres cultured in growth medium contained OCT4 and SOX2-positive cells distributed throughout the entire sphere, and no differentiating neurones. Gene expression of SOX2 and OCT4 mRNA detected by MBs correlated well with gene and protein expression measured by qRT-PCR and immunostaining, respectively. These experimental data support the theoretical model that stem cells cluster in the centre of neurospheres, and demonstrate the use of MBs for the spatial localization of specific gene-expressing cells within heterogeneous cell populations.
Collapse
Affiliation(s)
- Mirolyuba Ilieva
- Department of Micro- and Nanotechnology, Technical University of Denmark, Lyngby, Denmark
| | - Martin Dufva
- Department of Micro- and Nanotechnology, Technical University of Denmark, Lyngby, Denmark
- * E-mail:
| |
Collapse
|
34
|
Polarized neural stem cells derived from adult bone marrow stromal cells develop a rosette-like structure. In Vitro Cell Dev Biol Anim 2013; 49:638-52. [PMID: 23771792 DOI: 10.1007/s11626-013-9628-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 04/26/2013] [Indexed: 12/19/2022]
|
35
|
Abstract
Acute ischemic stroke causes a disturbance of neuronal circuitry and disruption of the blood-brain barrier that can lead to functional disabilities. At present, thrombolytic therapy inducing recanalization of the occluded vessels in the cerebral infarcted area is a commonly used therapeutic strategy. However, only a minority of patients have timely access to this kind of therapy. Recently, neural stem cells (NSCs) as therapy for stroke have been developed in preclinical studies. NSCs are harbored in the subventricular zone (SVZ) as well as the subgranular zone of the brain. The microenvironment in the SVZ, including intercellular interactions, extracellular matrix proteins, and soluble factors, can promote NSC proliferation, self-renewal, and multipotency. Endogenous neurogenesis responds to insults of ischemic stroke supporting the existence of remarkable plasticity in the mammalian brain. Homing and integration of NSCs to the sites of damaged brain tissue are complex morphological and physiological processes. This review provides an update on current preclinical cell therapies for stroke, focusing on neurogenesis in the SVZ and dentate gyrus and on recruitment cues that promote NSC homing and integration to the site of the damaged brain.
Collapse
Affiliation(s)
- Dah-Ching Ding
- Department of Obstetrics and Gynecology, Buddhist Tzu Chi General Hospital, Tzu Chi University, Hualien, Taiwan, ROC
| | | | | | | |
Collapse
|
36
|
Johnson NA, Slack GW, Savage KJ, Connors JM, Ben-Neriah S, Rogic S, Scott DW, Tan KL, Steidl C, Sehn LH, Chan WC, Iqbal J, Meyer PN, Lenz G, Wright G, Rimsza LM, Valentino C, Brunhoeber P, Grogan TM, Braziel RM, Cook JR, Tubbs RR, Weisenburger DD, Campo E, Rosenwald A, Ott G, Delabie J, Holcroft C, Jaffe ES, Staudt LM, Gascoyne RD. Concurrent expression of MYC and BCL2 in diffuse large B-cell lymphoma treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone. J Clin Oncol 2012; 30:3452-9. [PMID: 22851565 DOI: 10.1200/jco.2011.41.0985] [Citation(s) in RCA: 729] [Impact Index Per Article: 56.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
PURPOSE Diffuse large B-cell lymphoma (DLBCL) is curable in 60% of patients treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP). MYC translocations, with or without BCL2 translocations, have been associated with inferior survival in DLBCL. We investigated whether expression of MYC protein, with or without BCL2 protein expression, could risk-stratify patients at diagnosis. PATIENTS AND METHODS We determined the correlation between presence of MYC and BCL2 proteins by immunohistochemistry (IHC) with survival in two independent cohorts of patients with DLBCL treated with R-CHOP. We further determined if MYC protein expression correlated with high MYC mRNA and/or presence of MYC translocation. RESULTS In the training cohort (n = 167), MYC and BCL2 proteins were detected in 29% and 44% of patients, respectively. Concurrent expression (MYC positive/BCL2 positive) was present in 21% of patients. MYC protein correlated with presence of high MYC mRNA and MYC translocation (both P < .001), but the latter was less frequent (both 11%). MYC protein expression was only associated with inferior overall and progression-free survival when BCL2 protein was coexpressed (P < .001). Importantly, the poor prognostic effect of MYC positive/BCL2 positive was validated in an independent cohort of 140 patients with DLBCL and remained significant (P < .05) after adjusting for presence of high-risk features in a multivariable model that included elevated international prognostic index score, activated B-cell molecular subtype, and presence of concurrent MYC and BCL2 translocations. CONCLUSION Assessment of MYC and BCL2 expression by IHC represents a robust, rapid, and inexpensive approach to risk-stratify patients with DLBCL at diagnosis.
Collapse
|
37
|
Go HS, Kim KC, Choi CS, Jeon SJ, Kwon KJ, Han SH, Lee J, Cheong JH, Ryu JH, Kim CH, Ko KH, Shin CY. Prenatal exposure to valproic acid increases the neural progenitor cell pool and induces macrocephaly in rat brain via a mechanism involving the GSK-3β/β-catenin pathway. Neuropharmacology 2012; 63:1028-41. [PMID: 22841957 DOI: 10.1016/j.neuropharm.2012.07.028] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 07/11/2012] [Accepted: 07/18/2012] [Indexed: 12/01/2022]
Abstract
Autism is a spectrum of neurodevelopmental disorders characterized by social isolation and lack of interaction. Anatomically, autism patients often show macrocephaly and high neuronal density. To investigate the mechanism underlying the higher neuronal populations seen in ASD, we subcutaneously injected VPA (400 mg/kg) into pregnant Sprague-Dawley rats on E12, an animal model often used in ASD study. Alternatively, cultured rat neural progenitor cells were treated with VPA. Until E18, VPA induced NPC proliferation and delayed neurogenesis in fetal brain, but the subsequent differentiation of NPCs to neurons increased brain neuronal density afterward. Similar findings were observed with NPCs treated with VPA in vitro. At a molecular level, VPA enhanced Wnt1 expression and activated the GSK-3β/β-catenin pathway. Furthermore, inhibition of this pathway attenuated the effects of VPA. The findings of this study suggest that an altered developmental process underlies the macrocephaly and abnormal brain structure observed in the autistic brain.
Collapse
Affiliation(s)
- Hyo Sang Go
- Department of Pharmacology, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Cord blood-derived neuronal cells by ectopic expression of Sox2 and c-Myc. Proc Natl Acad Sci U S A 2012; 109:12556-61. [PMID: 22814375 DOI: 10.1073/pnas.1209523109] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The finding that certain somatic cells can be directly converted into cells of other lineages by the delivery of specific sets of transcription factors paves the way to novel therapeutic applications. Here we show that human cord blood (CB) CD133(+) cells lose their hematopoietic signature and are converted into CB-induced neuronal-like cells (CB-iNCs) by the ectopic expression of the transcription factor Sox2, a process that is further augmented by the combination of Sox2 and c-Myc. Gene-expression analysis, immunophenotyping, and electrophysiological analysis show that CB-iNCs acquire a distinct neuronal phenotype characterized by the expression of multiple neuronal markers. CB-iNCs show the ability to fire action potentials after in vitro maturation as well as after in vivo transplantation into the mouse hippocampus. This system highlights the potential of CB cells and offers an alternative means to the study of cellular plasticity, possibly in the context of drug screening research and of future cell-replacement therapies.
Collapse
|
39
|
Kerosuo L, Bronner-Fraser M. What is bad in cancer is good in the embryo: importance of EMT in neural crest development. Semin Cell Dev Biol 2012; 23:320-32. [PMID: 22430756 PMCID: PMC3345076 DOI: 10.1016/j.semcdb.2012.03.010] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Revised: 02/14/2012] [Accepted: 03/01/2012] [Indexed: 11/18/2022]
Abstract
Although the epithelial to mesenchymal transition (EMT) is famous for its role in cancer metastasis, it also is a normal developmental event in which epithelial cells are converted into migratory mesenchymal cells. A prime example of EMT during development occurs when neural crest (NC) cells emigrate from the neural tube thus providing an excellent model to study the principles of EMT in a nonmalignant environment. NC cells start life as neuroepithelial cells intermixed with precursors of the central nervous system. After EMT, they delaminate and begin migrating, often to distant sites in the embryo. While proliferating and maintaining multipotency and cell survival the transitioning neural crest cells lose apicobasal polarity and the basement membrane is broken down. This review discusses how these events are coordinated and regulated, by series of events involving signaling factors, gene regulatory interactions, as well as epigenetic and post-transcriptional modifications. Even though the series of events involved in NC EMT are well known, the sequence in which these steps take place remains a subject of debate, raising the intriguing possibility that, rather than being a single event, neural crest EMT may involve multiple parallel mechanisms.
Collapse
Affiliation(s)
- Laura Kerosuo
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, United States
| | | |
Collapse
|
40
|
Varlakhanova N, Cotterman R, Bradnam K, Korf I, Knoepfler PS. Myc and Miz-1 have coordinate genomic functions including targeting Hox genes in human embryonic stem cells. Epigenetics Chromatin 2011; 4:20. [PMID: 22053792 PMCID: PMC3226433 DOI: 10.1186/1756-8935-4-20] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 11/04/2011] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND A proposed role for Myc in maintaining mouse embryonic stem (ES) cell pluripotency is transcriptional repression of key differentiation-promoting genes, but detail of the mechanism has remained an important open topic. RESULTS To test the hypothesis that the zinc finger protein Miz-1 plays a central role, in the present work we conducted chromatin immunoprecipitation/microarray (ChIP-chip) analysis of Myc and Miz-1 in human ES cells, finding homeobox (Hox) genes as the most significant functional class of Miz-1 direct targets. Miz-1 differentiation-associated target genes specifically lack acetylated lysine 9 and trimethylated lysine 4 of histone H3 (AcH3K9 and H3K4me3) 9 histone marks, consistent with a repressed transcriptional state. Almost 30% of Miz-1 targets are also bound by Myc and these cobound genes are mostly factors that promote differentiation including Hox genes. Knockdown of Myc increased expression of differentiation genes directly bound by Myc and Miz-1, while a subset of the same genes is downregulated by Miz-1 loss-of-function. Myc and Miz-1 proteins interact with each other and associate with several corepressor factors in ES cells, suggesting a mechanism of repression of differentiation genes. CONCLUSIONS Taken together our data indicate that Miz-1 and Myc maintain human ES cell pluripotency by coordinately suppressing differentiation genes, particularly Hox genes. These data also support a new model of how Myc and Miz-1 function on chromatin.
Collapse
Affiliation(s)
- Natalia Varlakhanova
- Department of Cell Biology and Human Anatomy, University of California Davis School of Medicine, Sacramento, CA, USA.
| | | | | | | | | |
Collapse
|
41
|
Yamada K, Tso J, Ye F, Choe J, Liu Y, Liau LM, Tso CL. Essential gene pathways for glioblastoma stem cells: clinical implications for prevention of tumor recurrence. Cancers (Basel) 2011; 3:1975-95. [PMID: 24212792 PMCID: PMC3757400 DOI: 10.3390/cancers3021975] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2011] [Revised: 02/19/2011] [Accepted: 03/22/2011] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (World Health Organization/WHO grade IV) is the most common and most aggressive adult glial tumor. Patients with glioblastoma, despite being treated with gross total resection and post-operative radiation/chemotherapy, will almost always develop tumor recurrence. Glioblastoma stem cells (GSC), a minor subpopulation within the tumor mass, have been recently characterized as tumor-initiating cells and hypothesized to be responsible for post-treatment recurrence because of their enhanced radio-/chemo-resistant phenotype and ability to reconstitute tumors in mouse brains. Genome-wide expression profile analysis uncovered molecular properties of GSC distinct from their differentiated, proliferative progeny that comprise the majority of the tumor mass. In contrast to the hyperproliferative and hyperangiogenic phenotype of glioblastoma tumors, GSC possess neuroectodermal properties and express genes associated with neural stem cells, radial glial cells, and neural crest cells, as well as portray a migratory, quiescent, and undifferentiated phenotype. Thus, cell cycle-targeted radio-chemotherapy, which aims to kill fast-growing tumor cells, may not completely eliminate glioblastoma tumors. To prevent tumor recurrence, a strategy targeting essential gene pathways of GSC must be identified and incorporated into the standard treatment regimen. Identifying intrinsic and extrinsic cues by which GSC maintain stemness properties and sustain both tumorigenesis and anti-apoptotic features may provide new insights into potentially curative strategies for treating brain cancers.
Collapse
Affiliation(s)
- Kazunari Yamada
- Department of Surgery/Surgical Oncology, David Geffen School of Medicine, University of California Los Angeles, 13-260 Factor building, 10833 Le Conte Avenue, Los Angeles, California 90095, USA; E-Mails: (K.Y.); (J.T.); (F.Y.); (J.C.); (Y.L.); (C.L.T.)
| | - Jonathan Tso
- Department of Surgery/Surgical Oncology, David Geffen School of Medicine, University of California Los Angeles, 13-260 Factor building, 10833 Le Conte Avenue, Los Angeles, California 90095, USA; E-Mails: (K.Y.); (J.T.); (F.Y.); (J.C.); (Y.L.); (C.L.T.)
| | - Fei Ye
- Department of Surgery/Surgical Oncology, David Geffen School of Medicine, University of California Los Angeles, 13-260 Factor building, 10833 Le Conte Avenue, Los Angeles, California 90095, USA; E-Mails: (K.Y.); (J.T.); (F.Y.); (J.C.); (Y.L.); (C.L.T.)
| | - Jinny Choe
- Department of Surgery/Surgical Oncology, David Geffen School of Medicine, University of California Los Angeles, 13-260 Factor building, 10833 Le Conte Avenue, Los Angeles, California 90095, USA; E-Mails: (K.Y.); (J.T.); (F.Y.); (J.C.); (Y.L.); (C.L.T.)
| | - Yue Liu
- Department of Surgery/Surgical Oncology, David Geffen School of Medicine, University of California Los Angeles, 13-260 Factor building, 10833 Le Conte Avenue, Los Angeles, California 90095, USA; E-Mails: (K.Y.); (J.T.); (F.Y.); (J.C.); (Y.L.); (C.L.T.)
| | - Linda M. Liau
- Department of Neurosurgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA; E-Mail: (L.M.L.)
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, USA
| | - Cho-Lea Tso
- Department of Surgery/Surgical Oncology, David Geffen School of Medicine, University of California Los Angeles, 13-260 Factor building, 10833 Le Conte Avenue, Los Angeles, California 90095, USA; E-Mails: (K.Y.); (J.T.); (F.Y.); (J.C.); (Y.L.); (C.L.T.)
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-310-825-1066; Fax: +1- 310-825-7575
| |
Collapse
|
42
|
Wang C, Lisanti MP, Liao DJ. Reviewing once more the c-myc and Ras collaboration: converging at the cyclin D1-CDK4 complex and challenging basic concepts of cancer biology. Cell Cycle 2011; 10:57-67. [PMID: 21200143 DOI: 10.4161/cc.10.1.14449] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The c-myc is a proto-oncogene that manifests aberrant expression at high frequencies in most types of human cancer. C-myc gene amplifications are often observed in various cancers as well. Ample studies have also proved that c-myc has a potent oncogenicity, which can be further enhanced by collaborations with other oncogenes such as Bcl-2 and activated Ras. Studies on the collaborations of c-myc with Ras or other genes in oncogenicity have established several basic concepts and have disclosed their underlying mechanisms of tumor biology, including "immortalization" and "transformation". In many cases, these collaborations may converge at the cyclin D1-CDK4 complex. In the meantime, however, many results from studies on the c-myc, Ras and cyclin D1-CDK4 also challenge these basic concepts of tumor biology and suggest to us that the immortalized status of cells should be emphasized. Stricter criteria and definitions for a malignantly transformed status and a benign status of cells in culture also need to be established to facilitate our study of the mechanisms for tumor formation and to better link up in vitro data with animal results and eventually with human cancer pathology.
Collapse
Affiliation(s)
- Chenguang Wang
- Department of Stem Cell and Regenerative Medicine, and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | |
Collapse
|
43
|
Lee SH, Jeyapalan JN, Appleby V, Mohamed Noor DA, Sottile V, Scotting PJ. Dynamic methylation and expression of Oct4 in early neural stem cells. J Anat 2010; 217:203-13. [PMID: 20646110 DOI: 10.1111/j.1469-7580.2010.01269.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Neural stem cells are a multipotent population of tissue-specific stem cells with a broad but limited differentiation potential. However, recent studies have shown that over-expression of the pluripotency gene, Oct4, alone is sufficient to initiate a process by which these can form 'induced pluripotent stem cells' (iPS cells) with the same broad potential as embryonic stem cells. This led us to examine the expression of Oct4 in endogenous neural stem cells, as data regarding its expression in neural stem cells in vivo are contradictory and incomplete. In this study we have therefore analysed the expression of Oct4 and other genes associated with pluripotency throughout development of the mouse CNS and in neural stem cells grown in vitro. We find that Oct4 is still expressed in the CNS by E8.5, but that this expression declines rapidly until it is undetectable by E15.5. This decline is coincident with the gradual methylation of the Oct4 promoter and proximal enhancer. Immunostaining suggests that the Oct4 protein is predominantly cytoplasmic in location. We also found that neural stem cells from all ages expressed the pluripotency associated genes, Sox2, c-Myc, Klf4 and Nanog. These data provide an explanation for the varying behaviour of cells from the early neuroepithelium at different stages of development. The expression of these genes also provides an indication of why Oct4 alone is sufficient to induce iPS formation in neural stem cells at later stages.
Collapse
Affiliation(s)
- Shih-Han Lee
- Children's Brain Tumour Research Centre, Institute of Genetics, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | | | | | | | | | | |
Collapse
|