1
|
Han X, Jiang Z, Hou Y, Zhou X, Hu B. Myocardial ischemia-reperfusion injury upregulates nucleostemin expression via HIF-1α and c-Jun pathways and alleviates apoptosis by promoting autophagy. Cell Death Discov 2024; 10:461. [PMID: 39477962 PMCID: PMC11525682 DOI: 10.1038/s41420-024-02221-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 10/06/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
Myocardial ischemia-reperfusion (I/R) injury, often arising from interventional therapy for acute myocardial infarction, leads to irreversible myocardial cell death. While previous studies indicate that nucleostemin (NS) is induced by myocardial I/R injury and mitigates myocardial cell apoptosis, the underlying mechanisms are poorly understood. Here, our study reveals that NS upregulation is critical for preventing cardiomyocyte death following myocardial I/R injury. Elevated NS protein levels were observed in myocardial I/R injury mouse and rat models, as well as Hypoxia/reoxygenation (H/R) cardiac cell lines (H9C2 cells). We identified binding sites for c-Jun and HIF-1α in the NS promoter region. Inhibition of JNK and HIF-1α led to a significant decrease in NS transcription and protein expression. Furthermore, inhibition of autophagy and NS expression promoted myocardial cell apoptosis in H/R. Notably, the cell model showed reduced LC3I transformation to LC3II, downregulated Beclin1, upregulated p62, and altered expression of autophagy-related proteins upon NS interference in H/R cells. These findings suggest that NS expression, driven by c-Jun and HIF-1α pathways, facilitates autophagy, providing protection against both myocardial I/R injury and H/R-induced cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Xiao Han
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University & Department of Immunology, Medical School of Nantong University, Nantong, 226001, People's Republic of China
| | - Zhicheng Jiang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University & Department of Immunology, Medical School of Nantong University, Nantong, 226001, People's Republic of China
| | - Yufeng Hou
- Department of Immunology, Medical School of Nantong University, Nantong, 226001, People's Republic of China
| | - Xiaorong Zhou
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University & Department of Immunology, Medical School of Nantong University, Nantong, 226001, People's Republic of China.
- Department of Immunology, Medical School of Nantong University, Nantong, 226001, People's Republic of China.
| | - Baoying Hu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University & Department of Immunology, Medical School of Nantong University, Nantong, 226001, People's Republic of China.
- Department of Immunology, Medical School of Nantong University, Nantong, 226001, People's Republic of China.
| |
Collapse
|
2
|
Stepwise fate conversion of supporting cells to sensory hair cells in the chick auditory epithelium. iScience 2023; 26:106046. [PMID: 36818302 PMCID: PMC9932131 DOI: 10.1016/j.isci.2023.106046] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/17/2022] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
In contrast to mammals, the avian cochlea, specifically the basilar papilla, can regenerate sensory hair cells, which involves fate conversion of supporting cells to hair cells. To determine the mechanisms for converting supporting cells to hair cells, we used single-cell RNA sequencing during hair cell regeneration in explant cultures of chick basilar papillae. We identified dynamic changes in the gene expression of supporting cells, and the pseudotime trajectory analysis demonstrated the stepwise fate conversion from supporting cells to hair cells. Initially, supporting cell identity was erased and transition to the precursor state occurred. A subsequent gain in hair cell identity progressed together with downregulation of precursor-state genes. Transforming growth factor β receptor 1-mediated signaling was involved in induction of the initial step, and its inhibition resulted in suppression of hair cell regeneration. Our data provide new insights for understanding fate conversion from supporting cells to hair cells in avian basilar papillae.
Collapse
|
3
|
Yan D, Hua L. Nucleolar stress: Friend or foe in cardiac function? Front Cardiovasc Med 2022; 9:1045455. [PMID: 36386352 PMCID: PMC9659567 DOI: 10.3389/fcvm.2022.1045455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/17/2022] [Indexed: 03/14/2024] Open
Abstract
Studies in the past decades have uncovered an emerging role of the nucleolus in stress response and human disease progression. The disruption of ribosome biogenesis in the nucleolus causes aberrant nucleolar architecture and function, termed nucleolar stress, to initiate stress-responsive pathways via nucleolar release sequestration of various proteins. While data obtained from both clinical and basic investigations have faithfully demonstrated an involvement of nucleolar stress in the pathogenesis of cardiomyopathy, much remains unclear regarding its precise role in the progression of cardiac diseases. On the one hand, the initiation of nucleolar stress following acute myocardial damage leads to the upregulation of various cardioprotective nucleolar proteins, including nucleostemin (NS), nucleophosmin (NPM) and nucleolin (NCL). As a result, nucleolar stress plays an important role in facilitating the survival and repair of cardiomyocytes. On the other hand, abnormalities in nucleolar architecture and function are correlated with the deterioration of cardiac diseases. Notably, the cardiomyocytes of advanced ischemic and dilated cardiomyopathy display impaired silver-stained nucleolar organiser regions (AgNORs) and enlarged nucleoli, resembling the characteristics of tissue aging. Collectively, nucleolar abnormalities are critically involved in the development of cardiac diseases.
Collapse
Affiliation(s)
- Daliang Yan
- Department of Cardiovascular Surgery, Taizhou People’s Hospital, Taizhou, China
| | - Lu Hua
- Department of Oncology, Taizhou People’s Hospital, Taizhou, China
| |
Collapse
|
4
|
Quiroga-Artigas G, de Jong D, Schnitzler CE. GNL3 is an evolutionarily conserved stem cell gene influencing cell proliferation, animal growth and regeneration in the hydrozoan Hydractinia. Open Biol 2022; 12:220120. [PMID: 36069077 PMCID: PMC9449814 DOI: 10.1098/rsob.220120] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Nucleostemin (NS) is a vertebrate gene preferentially expressed in stem and cancer cells, which acts to regulate cell cycle progression, genome stability and ribosome biogenesis. NS and its paralogous gene, GNL3-like (GNL3L), arose in the vertebrate clade after a duplication event from their orthologous gene, G protein Nucleolar 3 (GNL3). Research on invertebrate GNL3, however, has been limited. To gain a greater understanding of the evolution and functions of the GNL3 gene, we have performed studies in the hydrozoan cnidarian Hydractinia symbiolongicarpus, a colonial hydroid that continuously generates pluripotent stem cells throughout its life cycle and presents impressive regenerative abilities. We show that Hydractinia GNL3 is expressed in stem and germline cells. The knockdown of GNL3 reduces the number of mitotic and S-phase cells in Hydractinia larvae of different ages. Genome editing of Hydractinia GNL3 via CRISPR/Cas9 resulted in colonies with reduced growth rates, polyps with impaired regeneration capabilities, gonadal morphological defects, and low sperm motility. Collectively, our study shows that GNL3 is an evolutionarily conserved stem cell and germline gene involved in cell proliferation, animal growth, regeneration and sexual reproduction in Hydractinia, and sheds new light into the evolution of GNL3 and of stem cell systems.
Collapse
Affiliation(s)
- Gonzalo Quiroga-Artigas
- Whitney Laboratory for Marine Bioscience, University of Florida, St Augustine, FL 32080, USA
| | - Danielle de Jong
- Whitney Laboratory for Marine Bioscience, University of Florida, St Augustine, FL 32080, USA
| | - Christine E Schnitzler
- Whitney Laboratory for Marine Bioscience, University of Florida, St Augustine, FL 32080, USA.,Department of Biology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
5
|
Cela I, Cufaro MC, Fucito M, Pieragostino D, Lanuti P, Sallese M, Del Boccio P, Di Matteo A, Allocati N, De Laurenzi V, Federici L. Proteomic Investigation of the Role of Nucleostemin in Nucleophosmin-Mutated OCI-AML 3 Cell Line. Int J Mol Sci 2022; 23:ijms23147655. [PMID: 35886999 PMCID: PMC9317519 DOI: 10.3390/ijms23147655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/07/2022] [Accepted: 07/09/2022] [Indexed: 02/06/2023] Open
Abstract
Nucleostemin (NS; a product of the GNL3 gene) is a nucleolar–nucleoplasm shuttling GTPase whose levels are high in stem cells and rapidly decrease upon differentiation. NS levels are also high in several solid and hematological neoplasms, including acute myeloid leukaemia (AML). While a role in telomere maintenance, response to stress stimuli and favoring DNA repair has been proposed in solid cancers, little or no information is available as to the role of nucleostemin in AML. Here, we investigate this issue via a proteomics approach. We use as a model system the OCI-AML 3 cell line harboring a heterozygous mutation at the NPM1 gene, which is the most frequent driver mutation in AML (approximately 30% of total AML cases). We show that NS is highly expressed in this cell line, and, contrary to what has previously been shown in other cancers, that its presence is dispensable for cell growth and viability. However, proteomics analysis of the OCI-AML 3 cell line before and after nucleostemin (NS) silencing showed several effects on different biological functions, as highlighted by ingenuity pathway analysis (IPA). In particular, we report an effect of down-regulating DNA repair through homologous recombination, and we confirmed a higher DNA damage rate in OCI-AML 3 cells when NS is depleted, which considerably increases upon stress induced by the topoisomerase II inhibitor etoposide. The data used are available via ProteomeXchange with the identifier PXD034012.
Collapse
Affiliation(s)
- Ilaria Cela
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (I.C.); (M.F.); (D.P.); (M.S.); (N.A.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.L.); (P.D.B.)
| | - Maria Concetta Cufaro
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.L.); (P.D.B.)
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Maurine Fucito
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (I.C.); (M.F.); (D.P.); (M.S.); (N.A.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.L.); (P.D.B.)
| | - Damiana Pieragostino
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (I.C.); (M.F.); (D.P.); (M.S.); (N.A.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.L.); (P.D.B.)
| | - Paola Lanuti
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.L.); (P.D.B.)
- Department of Medicine and Aging Science, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Michele Sallese
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (I.C.); (M.F.); (D.P.); (M.S.); (N.A.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.L.); (P.D.B.)
| | - Piero Del Boccio
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.L.); (P.D.B.)
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Adele Di Matteo
- Institute of Molecular Biology and Pathology, National Research Council of Italy, P.le Aldo Moro 5, 00185 Rome, Italy;
| | - Nerino Allocati
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (I.C.); (M.F.); (D.P.); (M.S.); (N.A.); (V.D.L.)
| | - Vincenzo De Laurenzi
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (I.C.); (M.F.); (D.P.); (M.S.); (N.A.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.L.); (P.D.B.)
| | - Luca Federici
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (I.C.); (M.F.); (D.P.); (M.S.); (N.A.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.L.); (P.D.B.)
- Correspondence:
| |
Collapse
|
6
|
Li J, Xu J, Wang Y, Li Q, Sun X, Fu W, Zhang B. Association of Nucleostemin Polymorphisms with Chronic Hepatitis B Virus Infection in Chinese Han Population. Genet Test Mol Biomarkers 2022; 26:255-262. [PMID: 35638911 PMCID: PMC9150128 DOI: 10.1089/gtmb.2021.0181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background: Chronic hepatitis B virus infection (CHB) is a common infectious disease that poses a global economic and health burden due to its high morbidity and mortality. Studies have demonstrated that host genetic factors play critical roles in the susceptibility and outcome of CHB. Aims: In this study, we aimed to assess the potential role of genetic variants of the nucleostemin (NS) gene with respect to CHB susceptibility. Materials and Methods: Four single nucleotide polymorphisms (SNPs) in the NS gene were genotyped in 446 patients with CHB and 399 healthy controls all of Chinese Han origin using the polymerase chain reaction-ligation detection reaction method. Results: The results showed that the three SNPs, rs3733039, rs1866268, and rs11177, were significantly associated with CHB. After a Bonferroni correction, the positive association of the rs3733039 SNP with CHB remained significant. Further analyses based on gender demonstrated that these SNPs are associated with CHB in both the female and male subgroups. After correction for multiple comparisons, all three SNPs in the female group were associated with CHB, whereas only the rs1866268 SNP in the male group was associated with CHB. Haplotype analysis showed that the C-C-G and T-T-T haplotypes in the block consisting of rs3733039-rs1866268-rs11177 were significantly associated with CHB. Conclusion: Our study demonstrated a genetic association between SNPs in the NS gene and the risk of CHB in the Chinese Han population for the first time. Thus, variations in the NS gene might serve as potential genetic biomarkers of CHB.
Collapse
Affiliation(s)
- Jixia Li
- Department of Clinical Laboratory, Yantaishan Hospital, Yantai, China
| | - Jinya Xu
- Department of Clinical Laboratory, Yantai Qishan Hospital, Yantai, China
| | - Yangui Wang
- Department of Clinical Laboratory, Yantaishan Hospital, Yantai, China
| | - Qin Li
- Department of Clinical Laboratory, Yantai Yuhuangding Hospital, Yantai, China
| | - Xilian Sun
- Department of Nursing, Yantaishan Hospital, Yantai, China
| | - Wen Fu
- Department of Clinical Laboratory, Yantaishan Hospital, Yantai, China
| | - Bo Zhang
- Department of Gastroenterology, Yantai Yuhuangding Hospital, Yantai, China
| |
Collapse
|
7
|
On the Cutting Edge of Oral Cancer Prevention: Finding Risk-Predictive Markers in Precancerous Lesions by Longitudinal Studies. Cells 2022; 11:cells11061033. [PMID: 35326482 PMCID: PMC8947091 DOI: 10.3390/cells11061033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/11/2022] [Accepted: 03/13/2022] [Indexed: 02/06/2023] Open
Abstract
Early identification and management of precancerous lesions at high risk of developing cancers is the most effective and economical way to reduce the incidence, mortality, and morbidity of cancers as well as minimizing treatment-related complications, including pain, impaired functions, and disfiguration. Reliable cancer-risk-predictive markers play an important role in enabling evidence-based decision making as well as providing mechanistic insight into the malignant conversion of precancerous lesions. The focus of this article is to review updates on markers that may predict the risk of oral premalignant lesions (OPLs) in developing into oral squamous cell carcinomas (OSCCs), which can logically be discovered only by prospective or retrospective longitudinal studies that analyze pre-progression OPL samples with long-term follow-up outcomes. These risk-predictive markers are different from those that prognosticate the survival outcome of cancers after they have been diagnosed and treated, or those that differentiate between different lesion types and stages. Up-to-date knowledge on cancer-risk-predictive markers discovered by longitudinally followed studies will be reviewed. The goal of this endeavor is to use this information as a starting point to address some key challenges limiting our progress in this area in the hope of achieving effective translation of research discoveries into new clinical interventions.
Collapse
|
8
|
Mobility of Nucleostemin in Live Cells Is Specifically Related to Transcription Inhibition by Actinomycin D and GTP-Binding Motif. Int J Mol Sci 2021; 22:ijms22158293. [PMID: 34361059 PMCID: PMC8347349 DOI: 10.3390/ijms22158293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/24/2021] [Accepted: 07/29/2021] [Indexed: 12/04/2022] Open
Abstract
In vertebrates, nucleostemin (NS) is an important marker of proliferation in several types of stem and cancer cells, and it can also interact with the tumor-suppressing transcription factor p53. In the present study, the intra-nuclear diffusional dynamics of native NS tagged with GFP and two GFP-tagged NS mutants with deleted guanosine triphosphate (GTP)-binding domains were analyzed by fluorescence correlation spectroscopy. Free and slow binding diffusion coefficients were evaluated, either under normal culture conditions or under treatment with specific cellular proliferation inhibitors actinomycin D (ActD), 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole (DRB), or trichostatin A (TSA). When treated with ActD, the fractional ratio of the slow diffusion was significantly decreased in the nucleoplasm. The decrease was proportional to ActD treatment duration. In contrast, DRB or TSA treatment did not affect NS diffusion. Interestingly, it was also found that the rate of diffusion of two NS mutants increased significantly even under normal conditions. These results suggest that the mobility of NS in the nucleoplasm is related to the initiation of DNA or RNA replication, and that the GTP-binding motif is also related to the large change of mobility.
Collapse
|
9
|
Li Y, Wu T, Liu S. Identification and Distinction of Tenocytes and Tendon-Derived Stem Cells. Front Cell Dev Biol 2021; 9:629515. [PMID: 33937230 PMCID: PMC8085586 DOI: 10.3389/fcell.2021.629515] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 03/29/2021] [Indexed: 01/01/2023] Open
Abstract
Restoring the normal structure and function of injured tendons is one of the biggest challenges in orthopedics and sports medicine department. The discovery of tendon-derived stem cells (TDSCs) provides a novel perspective to treat tendon injuries, which is expected to be an ideal seed cell to promote tendon repair and regeneration. Because of the lack of specific markers, the identification of tenocytes and TDSCs has not been conclusive in the in vitro study of tendons. In addition, the morphology of tendon derived cells is similar, and the comparison and identification of tenocytes and TDSCs are insufficient, which causes some obstacles to the in vitro study of tendon. In this review, the characteristics of tenocytes and TDSCs are summarized and compared based on some existing research results (mainly in terms of biomarkers), and a potential marker selection for identification is suggested. It is of profound significance to further explore the mechanism of biomarkers in vivo and to find more specific markers.
Collapse
Affiliation(s)
- Yuange Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tianyi Wu
- Department of Orthopaedics, Shanghai Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shen Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
10
|
Megiorni F, Camero S, Pontecorvi P, Camicia L, Marampon F, Ceccarelli S, Anastasiadou E, Bernabò N, Perniola G, Pizzuti A, Benedetti Panici P, Tombolini V, Marchese C. OTX015 Epi-Drug Exerts Antitumor Effects in Ovarian Cancer Cells by Blocking GNL3-Mediated Radioresistance Mechanisms: Cellular, Molecular and Computational Evidence. Cancers (Basel) 2021; 13:cancers13071519. [PMID: 33806232 PMCID: PMC8059141 DOI: 10.3390/cancers13071519] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/10/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The outcome for women diagnosed with ovarian cancer (OC), the most aggressive gynecological tumor worldwide, remains very poor. Encouraging therapeutic impact of epigenetic drugs has been suggested in a wide range of human solid tumors, including OC. The present study assessed the in vitro cytostatic and cytotoxic effects of OTX015, a pan Bromodomain and Extra-Terminal motif inhibitor, in human OC cells, both as single treatment and in combination with radiotherapy. Cellular, molecular and computational network analyses indicated the centrality of GNL3 downregulation in mediating the OTX015-related antitumor efficacy that blocks disease progression/maintenance and radioresistance acquisition. Our preclinical results confirm that targeted and combinatorial treatments represent effective anticancer strategies to be translated in the clinical research for improving OC patient care. Abstract Ovarian cancer (OC) is the most aggressive gynecological tumor worldwide and, notwithstanding the increment in conventional treatments, many resistance mechanisms arise, this leading to cure failure and patient death. So, the use of novel adjuvant drugs able to counteract these pathways is urgently needed to improve patient overall survival. A growing interest is focused on epigenetic drugs for cancer therapy, such as Bromodomain and Extra-Terminal motif inhibitors (BETi). Here, we investigate the antitumor effects of OTX015, a novel BETi, as a single agent or in combination with ionizing radiation (IR) in OC cellular models. OTX015 treatment significantly reduced tumor cell proliferation by triggering cell cycle arrest and apoptosis that were linked to nucleolar stress and DNA damage. OTX015 impaired migration capacity and potentiated IR effects by reducing the expression of different drivers of cancer resistance mechanisms, including GNL3 gene, whose expression was found to be significantly higher in OC biopsies than in normal ovarian tissues. Gene specific knocking down and computational network analysis confirmed the centrality of GNL3 in OTX015-mediated OC antitumor effects. Altogether, our findings suggest OTX015 as an effective option to improve therapeutic strategies and overcome the development of resistant cancer cells in patients with OC.
Collapse
Affiliation(s)
- Francesca Megiorni
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (E.A.); (A.P.); (C.M.)
- Correspondence: ; Tel.: +39-06-4997-8272
| | - Simona Camero
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (S.C.); (L.C.); (G.P.); (P.B.P.)
| | - Paola Pontecorvi
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (E.A.); (A.P.); (C.M.)
| | - Lucrezia Camicia
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (S.C.); (L.C.); (G.P.); (P.B.P.)
| | - Francesco Marampon
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (F.M.); (V.T.)
| | - Simona Ceccarelli
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (E.A.); (A.P.); (C.M.)
| | - Eleni Anastasiadou
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (E.A.); (A.P.); (C.M.)
| | - Nicola Bernabò
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy;
| | - Giorgia Perniola
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (S.C.); (L.C.); (G.P.); (P.B.P.)
| | - Antonio Pizzuti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (E.A.); (A.P.); (C.M.)
| | - Pierluigi Benedetti Panici
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (S.C.); (L.C.); (G.P.); (P.B.P.)
| | - Vincenzo Tombolini
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (F.M.); (V.T.)
| | - Cinzia Marchese
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (E.A.); (A.P.); (C.M.)
| |
Collapse
|
11
|
Wong CY, Chang YM, Tsai YS, Ng WV, Cheong SK, Chang TY, Chung IF, Lim YM. Decoding the differentiation of mesenchymal stem cells into mesangial cells at the transcriptomic level. BMC Genomics 2020; 21:467. [PMID: 32635896 PMCID: PMC7339572 DOI: 10.1186/s12864-020-06868-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 06/23/2020] [Indexed: 02/08/2023] Open
Abstract
Background Mesangial cells play an important role in the glomerulus to provide mechanical support and maintaine efficient ultrafiltration of renal plasma. Loss of mesangial cells due to pathologic conditions may lead to impaired renal function. Mesenchymal stem cells (MSC) can differentiate into many cell types, including mesangial cells. However transcriptomic profiling during MSC differentiation into mesangial cells had not been studied yet. The aim of this study is to examine the pattern of transcriptomic changes during MSC differentiation into mesangial cells, to understand the involvement of transcription factor (TF) along the differentiation process, and finally to elucidate the relationship among TF-TF and TF-key gene or biomarkers during the differentiation of MSC into mesangial cells. Results Several ascending and descending monotonic key genes were identified by Monotonic Feature Selector. The identified descending monotonic key genes are related to stemness or regulation of cell cycle while ascending monotonic key genes are associated with the functions of mesangial cells. The TFs were arranged in a co-expression network in order of time by Time-Ordered Gene Co-expression Network (TO-GCN) analysis. TO-GCN analysis can classify the differentiation process into three stages: differentiation preparation, differentiation initiation and maturation. Furthermore, it can also explore TF-TF-key genes regulatory relationships in the muscle contraction process. Conclusions A systematic analysis for transcriptomic profiling of MSC differentiation into mesangial cells has been established. Key genes or biomarkers, TFs and pathways involved in differentiation of MSC-mesangial cells have been identified and the related biological implications have been discussed. Finally, we further elucidated for the first time the three main stages of mesangial cell differentiation, and the regulatory relationships between TF-TF-key genes involved in the muscle contraction process. Through this study, we have increased fundamental understanding of the gene transcripts during the differentiation of MSC into mesangial cells.
Collapse
Affiliation(s)
- Chee-Yin Wong
- Department of Pre-Clinical Sciences, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Jalan Sungai Long, Bandar Sungai Long, 43000 Kajang, Selangor, Malaysia
| | - Yao-Ming Chang
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Yu-Shuen Tsai
- Center for Systems and Synthetic Biology, National Yang-Ming University, No. 155, Section 2, Linong Street, Taipei, Taiwan
| | - Wailap Victor Ng
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, No. 155, Section 2, Linong Street, Taipei, Taiwan
| | - Soon-Keng Cheong
- Department of Pre-Clinical Sciences, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Jalan Sungai Long, Bandar Sungai Long, 43000 Kajang, Selangor, Malaysia
| | - Ting-Yu Chang
- Department of Research, ChangHua Christian Hospital, 135, Nan-Hsiao Street, ChangHua City, Taiwan
| | - I-Fang Chung
- Center for Systems and Synthetic Biology, National Yang-Ming University, No. 155, Section 2, Linong Street, Taipei, Taiwan. .,Institute of Biomedical Informatics, National Yang-Ming University, No. 155, Section 2, Linong Street, Taipei, Taiwan. .,Preventive Medicine Research Center, National Yang-Ming University, No. 155, Section 2, Linong Street, Taipei, Taiwan.
| | - Yang-Mooi Lim
- Department of Pre-Clinical Sciences, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Jalan Sungai Long, Bandar Sungai Long, 43000 Kajang, Selangor, Malaysia.
| |
Collapse
|
12
|
Wang J, McGrail DJ, Bhupal PK, Zhang W, Lin KY, Ku YH, Lin T, Wu H, Tsai KC, Li K, Peng CY, Finegold MJ, Lin SY, Tsai RYL. Nucleostemin Modulates Outcomes of Hepatocellular Carcinoma via a Tumor Adaptive Mechanism to Genomic Stress. Mol Cancer Res 2020; 18:723-734. [PMID: 32051231 PMCID: PMC7202947 DOI: 10.1158/1541-7786.mcr-19-0777] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 01/03/2020] [Accepted: 02/05/2020] [Indexed: 12/14/2022]
Abstract
Hepatocellular carcinomas (HCC) are adapted to survive extreme genomic stress conditions imposed by hyperactive DNA replication and genotoxic drug treatment. The underlying mechanisms remain unclear, but may involve intensified DNA damage response/repair programs. Here, we investigate a new role of nucleostemin (NS) in allowing HCC to survive its own malignancy, as NS was previously shown to promote liver regeneration via a damage repair mechanism. We first established that a higher NS transcript level correlates with high-HCC grades and poor prognostic signatures, and is an independent predictor of shorter overall and progression-free survival specifically for HCC and kidney cancer but not for others. Immunostaining confirmed that NS is most abundantly expressed in high-grade and metastatic HCCs. Genome-wide analyses revealed that NS is coenriched with MYC target and homologous recombination (HR) repair genes in human HCC samples and functionally intersects with those involved in replication stress response and HR repair in yeasts. In support, NS-high HCCs are more reliant on the replicative/oxidative stress response pathways, whereas NS-low HCCs depend more on the mTOR pathway. Perturbation studies showed NS function in protecting human HCC cells from replication- and drug-induced DNA damage. Notably, NS depletion in HCC cells increases the amounts of physical DNA damage and cytosolic double-stranded DNA, leading to a reactive increase of cytokines and PD-L1. This study shows that NS provides an essential mechanism for HCC to adapt to high genomic stress for oncogenic maintenance and propagation. NS deficiency sensitizes HCC cells to chemotherapy but also triggers tumor immune responses. IMPLICATIONS: HCC employs a novel, nucleostemin (NS)-mediated-mediated adaptive mechanism to survive high genomic stress conditions, a deficiency of which sensitizes HCC cells to chemotherapy but also triggers tumor immune responses.
Collapse
Affiliation(s)
- Junying Wang
- Institute of Biosciences and Technology, Texas A&M University, Houston, Texas
| | - Daniel J McGrail
- Department of Systems Biology, MD Anderson Cancer Center, Houston, Texas
| | - Parnit K Bhupal
- Institute of Biosciences and Technology, Texas A&M University, Houston, Texas
| | - Wen Zhang
- Institute of Biosciences and Technology, Texas A&M University, Houston, Texas
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kuan-Yu Lin
- Institute of Biosciences and Technology, Texas A&M University, Houston, Texas
| | - Yi-Hsuan Ku
- Institute of Biosciences and Technology, Texas A&M University, Houston, Texas
| | - Tao Lin
- Institute of Biosciences and Technology, Texas A&M University, Houston, Texas
| | - Hongfu Wu
- Institute of Biosciences and Technology, Texas A&M University, Houston, Texas
| | - Kyle C Tsai
- Michael E. DeBakey High School for Health Professions, Houston, Texas
| | - Kaiyi Li
- Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Cheng-Yuan Peng
- Division of Hepatogastroenterology, Department of Internal Medicine, China Medical University Hospital, School of Medicine, China Medical University, Taichung, Taiwan
| | - Milton J Finegold
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas
| | - Shiaw-Yih Lin
- Department of Systems Biology, MD Anderson Cancer Center, Houston, Texas
| | - Robert Y L Tsai
- Institute of Biosciences and Technology, Texas A&M University, Houston, Texas.
| |
Collapse
|
13
|
Camero S, Camicia L, Marampon F, Ceccarelli S, Shukla R, Mannarino O, Pizer B, Schiavetti A, Pizzuti A, Tombolini V, Marchese C, Dominici C, Megiorni F. BET inhibition therapy counteracts cancer cell survival, clonogenic potential and radioresistance mechanisms in rhabdomyosarcoma cells. Cancer Lett 2020; 479:71-88. [PMID: 32200036 DOI: 10.1016/j.canlet.2020.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/04/2020] [Accepted: 03/10/2020] [Indexed: 12/15/2022]
Abstract
The antitumour effects of OTX015, a first-in-class BET inhibitor (BETi), were investigated as a single agent or in combination with ionizing radiation (IR) in preclinical in vitro models of rhabdomyosarcoma (RMS), the most common childhood soft tissue sarcoma. Herein, we demonstrated the upregulation of BET Bromodomain gene expression in RMS tumour biopsies and cell lines compared to normal skeletal muscle. In vitro experiments showed that OTX015 significantly reduced RMS cell proliferation by altering cell cycle modulators and apoptotic related proteins due to the accumulation of DNA breaks that cells are unable to repair. Interestingly, OTX015 also impaired migration capacity and tumour-sphere architecture by downregulating pro-stemness genes and was able to potentiate ionizing radiation effects by reducing the expression of different drivers of tumour dissemination and resistance mechanisms, including the GNL3 gene, that we correlated for the first time with the RMS phenotype. In conclusion, our research sheds further light on the molecular events of OTX015 action against RMS cells and indicates this novel BETi as an effective option to improve therapeutic strategies and overcome the development of resistant cancer cells in patients with RMS.
Collapse
Affiliation(s)
- Simona Camero
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Lucrezia Camicia
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Francesco Marampon
- Department of Radiological, Oncological and Pathological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Simona Ceccarelli
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| | - Rajeev Shukla
- Department of Perinatal and Paediatric Pathology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK.
| | - Olga Mannarino
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Barry Pizer
- Department of Oncology, Alder Hey Children's NHS Foundation Trust, Eaton Road, Liverpool, L12 2AP, UK.
| | - Amalia Schiavetti
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Antonio Pizzuti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| | - Vincenzo Tombolini
- Department of Radiological, Oncological and Pathological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Cinzia Marchese
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| | - Carlo Dominici
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Francesca Megiorni
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| |
Collapse
|
14
|
Ribosome and Translational Control in Stem Cells. Cells 2020; 9:cells9020497. [PMID: 32098201 PMCID: PMC7072746 DOI: 10.3390/cells9020497] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/11/2020] [Accepted: 02/17/2020] [Indexed: 12/16/2022] Open
Abstract
Embryonic stem cells (ESCs) and adult stem cells (ASCs) possess the remarkable capacity to self-renew while remaining poised to differentiate into multiple progenies in the context of a rapidly developing embryo or in steady-state tissues, respectively. This ability is controlled by complex genetic programs, which are dynamically orchestrated at different steps of gene expression, including chromatin remodeling, mRNA transcription, processing, and stability. In addition to maintaining stem cell homeostasis, these molecular processes need to be rapidly rewired to coordinate complex physiological modifications required to redirect cell fate in response to environmental clues, such as differentiation signals or tissue injuries. Although chromatin remodeling and mRNA expression have been extensively studied in stem cells, accumulating evidence suggests that stem cell transcriptomes and proteomes are poorly correlated and that stem cell properties require finely tuned protein synthesis. In addition, many studies have shown that the biogenesis of the translation machinery, the ribosome, is decisive for sustaining ESC and ASC properties. Therefore, these observations emphasize the importance of translational control in stem cell homeostasis and fate decisions. In this review, we will provide the most recent literature describing how ribosome biogenesis and translational control regulate stem cell functions and are crucial for accommodating proteome remodeling in response to changes in stem cell fate.
Collapse
|
15
|
Identification and Characterization of the Nucleolar Localization Signal of Autographa californica Multiple Nucleopolyhedrovirus LEF5. J Virol 2020; 94:JVI.01891-19. [PMID: 31776271 DOI: 10.1128/jvi.01891-19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 11/19/2019] [Indexed: 11/20/2022] Open
Abstract
Autographa californica multiple nucleopolyhedrovirus (AcMNPV) late expression factor 5 (LEF5) is highly conserved in all sequenced baculovirus genomes and plays an important role in production of infectious viral progeny. In this study, nucleolar localization of AcMNPV LEF5 was characterized. Through transcriptome analysis, we identified two putative nucleolar proteins, Spodoptera frugiperda nucleostemin (SfNS) and fibrillarin (SfFBL), from Sf9 cells. Immunofluorescence analysis demonstrated that SfNS and SfFBL were localized to the nucleolus. AcMNPV infection resulted in reorganization of the nucleoli of infected cells. Colocalization of LEF5 and SfNS showed that AcMNPV LEF5 was localized to the nucleolus in Sf9 cells. Bioinformatic analysis revealed that basic amino acids of LEF5 are enriched at residues 184 to 213 and may contain a nucleolar localization signal (NoLS). Green fluorescent protein (GFP) fused to NoLS of AcMNPV LEF5 localized to the nucleoli of transfected cells. Multiple-point mutation analysis demonstrated that amino acid residues 197 to 204 are important for nucleolar localization of LEF5. To identify whether the NoLS in AcMNPV LEF5 is important for production of viral progeny, a lef5-null AcMNPV bacmid was constructed; several NoLS-mutated LEF5 proteins were reinserted into the lef5-null AcMNPV bacmid with a GFP reporter. The constructs containing point mutations at residues 185 to 189 or 197 to 204 in AcMNPV LEF5 resulted in reduction in production of infectious viral progeny and occlusion body yield in bacmid-transfected cells. Together, these data suggested that AcMNPV LEF5 contains an NoLS, which is important for nucleolar localization of LEF5, progeny production, and occlusion body production.IMPORTANCE Many viruses, including human and plant viruses, target nucleolar functions as part of their infection strategy. However, nucleolar localization for baculovirus proteins has not yet been characterized. In this study, two nucleolar proteins, SfNS and SfFBL, were identified in Sf9 cells. Our results showed that Autographa californica multiple nucleopolyhedrovirus (AcMNPV) infection resulted in redistribution of the nucleoli of infected cells. We demonstrated that AcMNPV late expression factor 5 (LEF5) could localize to the nucleolus and contains a nucleolar localization signal (NoLS), which is important for nucleolar localization of AcMNPV LEF5 and for production of viral progeny and yield of occlusion bodies.
Collapse
|
16
|
Saito T. A Nucleolar Protein, Nepro, Is Essential for the Maintenance of Early Neural Stem Cells and Preimplantation Embryos. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1218:93-101. [PMID: 32060873 DOI: 10.1007/978-3-030-34436-8_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Notch signaling is required for maintaining neural stem cells (NSCs) in the developing brain. NSCs have potential to give rise to many neuronal types in the early telencephalon, and the potential decreases as embryonic development proceeds. Nepro, which encodes a unique nucleolar protein and is activated downstream of Notch, is essential for maintaining NSCs in the early telencephalon. Nepro is also expressed at basal levels and required for maintaining the preimplantation embryo, by repressing mitochondria-associated p53 apoptotic signaling. Notch signaling also controls dendritic complexity in mitral cells, major projection neurons in the olfactory bulb, showing that many steps of neural development involve Notch signaling.
Collapse
Affiliation(s)
- Tetsuichiro Saito
- Department of Developmental Biology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| |
Collapse
|
17
|
Sami MM, Hachim MY, Hachim IY, Elbarkouky AH, López-Ozuna VM. Nucleostemin expression in breast cancer is a marker of more aggressive phenotype and unfavorable patients' outcome: A STROBE-compliant article. Medicine (Baltimore) 2019; 98:e14744. [PMID: 30817632 PMCID: PMC6831441 DOI: 10.1097/md.0000000000014744] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Cancer stem cells (CSCs) are postulated to play significant role in the pathogenesis, progression as well as drug resistance of breast cancer. Nucleostemin (NS) is thought to be a key molecule for stemness, and the clinical impact of NS immunoreactivity in breast cancer can indicate its actual role and future therapeutic potentials.The current study is an observational study with an attempt to evaluate the correlation between NS expression (protein and gene expression levels) and different clinicopathological attributes of invasive breast cancer. For that reason, we investigated NS immunohistochemistry expression on commercial tissue microarray (TMA) of 102 patients and 51 archival specimens from patients admitted to Saqr Hospital, Ras Al Khaimah and diagnosed in Al Baraha Hospital, Dubai, UAE. In addition, the association between NS (GNL3) gene expression and different prognostic parameters as well as patient outcome was also evaluated using 2 large publicly available databases.Interestingly, we found NS expression to be associated with less differentiated and more advance stage. In addition, NS expression was significantly higher in larger size (P = .001) and LN-positive tumors (P = .007). Notably, NS expression was significantly correlated to P53 positive (P = .037) status. Furthermore, NS was found to be more expressed in the highly aggressive breast cancer subtypes including human epidermal growth factor receptor 2 (HER-2) and triple negative breast cancer (TNBC) subtypes. Moreover, our results also showed that high GNL3 gene expression to be associated with poor patient outcome and higher chances of tumor recurrence.Our results highlight NS expression as a marker of aggressive phenotype and poor outcome and indicate its possible use as a potential target for CSC-associated breast cancer management.
Collapse
Affiliation(s)
- Manal M. Sami
- Department of Pathology, Ras Al Khaimah College of Medical Sciences, Ras Al Khaimah Medical and Health Sciences University, United Arab Emirates
- Department of Pathology, Faculty of Medicine, Suez Canal University, Ismaillia, Egypt
| | - Mahmood Y. Hachim
- Sharjah Institute for Medical Research, University of Sharjah, United Arab Emirates
| | - Ibrahim Y. Hachim
- Department of Medicine, McGill University Health Center, Cancer Research Program, Montreal, Canada
| | - Ahmed H. Elbarkouky
- Department of Pathology, Al Baraha Hospital, Dubai, United Arab Emirates
- Department of Pathology, College of Medicine, Tanta University, Tanta, Gharbia, Egypt
| | - Vanessa M. López-Ozuna
- Segal Cancer Center, Lady Davis Institute of Medical Research, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
18
|
Lin T, Lin TC, McGrail DJ, Bhupal PK, Ku YH, Zhang W, Meng L, Lin SY, Peng G, Tsai RYL. Nucleostemin reveals a dichotomous nature of genome maintenance in mammary tumor progression. Oncogene 2019; 38:3919-3931. [PMID: 30692636 PMCID: PMC6525051 DOI: 10.1038/s41388-019-0710-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 01/11/2019] [Accepted: 01/15/2019] [Indexed: 12/17/2022]
Abstract
A defective homologous recombination (HR) repair program increases tumor incidence as well as providing a survival advantage in patients with breast and ovarian cancers. Here, we hypothesize that the tumor-promoting side of genome maintenance programs may be contributed by a self-renewal protein, nucleostemin (NS). To address this issue, we established its functional importance in mammary tumor progression in mice and showed that mammary tumor cells become highly susceptible to replicative DNA damage following NS depletion and are protected from hydroxyurea-induced damage by NS overexpression. Breast cancer cells with basal-like characters display more reliance on NS for genome maintenance than those with luminal characters. Mechanistically, NS-deficient cells demonstrate a significantly reduced HR repair activity. TCGA analyses of human breast cancers revealed that NS is co-enriched positively with HR repair proteins and that high NS expression correlates with low HR defects and predicts poor progression-free survival and resistance to knockdown of cell cycle checkpoint genes in triple-negative/basal-like breast cancers. This work indicates that NS constitutes a tumor-promoting genome maintenance program required for mammary tumor progression.
Collapse
Affiliation(s)
- Tao Lin
- Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Tsung-Chin Lin
- Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Daniel J McGrail
- Department of Systems Biology, MD Anderson Cancer Center, Houston, TX, USA
| | - Parnit K Bhupal
- Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Yi-Hsuan Ku
- Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Wen Zhang
- Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Lingjun Meng
- Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Shiaw-Yih Lin
- Department of Systems Biology, MD Anderson Cancer Center, Houston, TX, USA
| | - Guang Peng
- Department of Clinical Cancer Prevention, MD Anderson Cancer Center, Houston, TX, USA
| | - Robert Y L Tsai
- Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA. .,Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
19
|
Bono B, Ostano P, Peritore M, Gregnanin I, Belgiovine C, Liguori M, Allavena P, Chiorino G, Chiodi I, Mondello C. Cells with stemness features are generated from in vitro transformed human fibroblasts. Sci Rep 2018; 8:13838. [PMID: 30218041 PMCID: PMC6138721 DOI: 10.1038/s41598-018-32197-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 08/30/2018] [Indexed: 12/19/2022] Open
Abstract
Cancer stem cells (CSCs) have been involved in the maintenance, progression and relapse of several tumors, but their origin is still elusive. Here, in vitro transformed human fibroblasts (cen3tel cells) and the tumorsphere assay were used to search for and possibly characterize CSCs in transformed somatic cells. Cen3tel cells formed spheres showing self-renewal capacity and Sox2 overexpression, suggesting that they contained a subset of cells with CSC-like features. Sphere cells displayed deregulation of a c-MYC/miR-34a circuitry, likely associated with cell protection from apoptosis. Gene expression profiles of sphere cells revealed an extensive transcriptional reprogramming. Genes up-regulated in tumorspheres identified processes related to tumorigenesis and stemness, as cholesterol biosynthesis, apoptosis suppression, interferon and cytokine mediated signalling pathways. Sphere cells engrafted into NSG mice more rapidly than adherent cells, but both cell populations were tumorigenic. These results indicate that, during transformation, human somatic cells can acquire CSC properties, confirming the high plasticity of tumor cells. However, CSC-like cells are not the only tumorigenic population in transformed cells, indicating that the CSC phenotype and tumorigenicity can be uncoupled.
Collapse
Affiliation(s)
- Bartolo Bono
- Istituto di Genetica Molecolare, CNR, Via Abbiategrasso, 207, 27100, Pavia, Italy.,Dipartimento di Biologia e Biotecnologie, Pavia University, via Ferrata 9, 27100, Pavia, Italy
| | - Paola Ostano
- "Cancer Genomics Laboratory" Fondazione Edo ed Elvo Tempia Valenta, Via Malta, 3, 13900, Biella, Italy
| | - Martina Peritore
- Istituto di Genetica Molecolare, CNR, Via Abbiategrasso, 207, 27100, Pavia, Italy
| | - Ilaria Gregnanin
- "Cancer Genomics Laboratory" Fondazione Edo ed Elvo Tempia Valenta, Via Malta, 3, 13900, Biella, Italy
| | - Cristina Belgiovine
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 20090, Pieve Emanuele, Milan, Italy
| | - Manuela Liguori
- IRCCS Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Paola Allavena
- IRCCS Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Giovanna Chiorino
- "Cancer Genomics Laboratory" Fondazione Edo ed Elvo Tempia Valenta, Via Malta, 3, 13900, Biella, Italy
| | - Ilaria Chiodi
- Istituto di Genetica Molecolare, CNR, Via Abbiategrasso, 207, 27100, Pavia, Italy.
| | - Chiara Mondello
- Istituto di Genetica Molecolare, CNR, Via Abbiategrasso, 207, 27100, Pavia, Italy.
| |
Collapse
|
20
|
Ewing RM, Song J, Gokulrangan G, Bai S, Bowler EH, Bolton R, Skipp P, Wang Y, Wang Z. Multiproteomic and Transcriptomic Analysis of Oncogenic β-Catenin Molecular Networks. J Proteome Res 2018; 17:2216-2225. [PMID: 29747501 DOI: 10.1021/acs.jproteome.8b00180] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The dysregulation of Wnt signaling is a frequent occurrence in many different cancers. Oncogenic mutations of CTNNB1/β-catenin, the key nuclear effector of canonical Wnt signaling, lead to the accumulation and stabilization of β-catenin protein with diverse effects in cancer cells. Although the transcriptional response to Wnt/β-catenin signaling activation has been widely studied, an integrated understanding of the effects of oncogenic β-catenin on molecular networks is lacking. We used affinity-purification mass spectrometry (AP-MS), label-free liquid chromatography-tandem mass spectrometry, and RNA-Seq to compare protein-protein interactions, protein expression, and gene expression in colorectal cancer cells expressing mutant (oncogenic) or wild-type β-catenin. We generate an integrated molecular network and use it to identify novel protein modules that are associated with mutant or wild-type β-catenin. We identify a DNA methyltransferase I associated subnetwork that is enriched in cells with mutant β-catenin and a subnetwork enriched in wild-type cells associated with the CDKN2A tumor suppressor, linking these processes to the transformation of colorectal cancer cells through oncogenic β-catenin signaling. In summary, multiomics analysis of a defined colorectal cancer cell model provides a significantly more comprehensive identification of functional molecular networks associated with oncogenic β-catenin signaling.
Collapse
Affiliation(s)
- Rob M Ewing
- School of Biological Sciences , University of Southampton , Southampton SO17 1BJ , United Kingdom
| | - Jing Song
- School of Medicine , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| | - Giridharan Gokulrangan
- School of Medicine , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| | - Sheldon Bai
- School of Medicine , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| | - Emily H Bowler
- School of Biological Sciences , University of Southampton , Southampton SO17 1BJ , United Kingdom
| | - Rachel Bolton
- School of Biological Sciences , University of Southampton , Southampton SO17 1BJ , United Kingdom
| | - Paul Skipp
- School of Biological Sciences , University of Southampton , Southampton SO17 1BJ , United Kingdom
| | - Yihua Wang
- School of Biological Sciences , University of Southampton , Southampton SO17 1BJ , United Kingdom
| | - Zhenghe Wang
- School of Medicine , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| |
Collapse
|
21
|
Abstract
Most of the adult CNS lacks regenerative activity in terms of both neuron birth and neurite outgrowth. While this regeneration-unfriendly environment of the adult CNS may preserve the existing neuronal circuitry that takes years to develop in higher organisms, it also poses a major obstacle for CNS repair later in life. In this issue of the JCI, Song et al. report on their development of a strategy that uses region-specific and molecularly engineered astrocytes to turn an unfavorable brain environment into a favorable one for engrafted neural stem/progenitor cells (NSC/NPCs). In a rat model of Parkinson's disease (PD), cografting NPCs with midbrain-derived astrocytes engineered to overexpress the transcription factors Nurr1 and Foxa2 promotes maturation and survival of the graft, resulting in therapeutic improvement. The results of this study raise the prospect of using modified astrocytes to improve the survival, maturation, and integration of engrafted NSC/NPCs as a restorative treatment for PD.
Collapse
|
22
|
Schwalie PC, Ordóñez-Morán P, Huelsken J, Deplancke B. Cross-Tissue Identification of Somatic Stem and Progenitor Cells Using a Single-Cell RNA-Sequencing Derived Gene Signature. Stem Cells 2017; 35:2390-2402. [PMID: 29044933 DOI: 10.1002/stem.2719] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 09/27/2017] [Accepted: 10/02/2017] [Indexed: 12/15/2022]
Abstract
A long-standing question in biology is whether multipotent somatic stem and progenitor cells (SSPCs) feature molecular properties that could guide their system-independent identification. Population-based transcriptomic studies have so far not been able to provide a definite answer, given the rarity and heterogeneous nature of these cells. Here, we exploited the resolving power of single-cell RNA-sequencing to develop a computational model that is able to accurately distinguish SSPCs from differentiated cells across tissues. The resulting classifier is based on the combined expression of 23 genes including known players in multipotency, proliferation, and tumorigenesis, as well as novel ones, such as Lcp1 and Vgll4 that we functionally validate in intestinal organoids. We show how this approach enables the identification of stem-like cells in still ambiguous systems such as the pancreas and the epidermis as well as the exploration of lineage commitment hierarchies, thus facilitating the study of biological processes such as cellular differentiation, tissue regeneration, and cancer. Stem Cells 2017;35:2390-2402.
Collapse
Affiliation(s)
- Petra C Schwalie
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering and Swiss Institute of Bioinformatics, CH-1015, Lausanne, Switzerland
| | - Paloma Ordóñez-Morán
- ISREC (Swiss Institute for Experimental Cancer Research), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Joerg Huelsken
- ISREC (Swiss Institute for Experimental Cancer Research), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Bart Deplancke
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering and Swiss Institute of Bioinformatics, CH-1015, Lausanne, Switzerland
| |
Collapse
|
23
|
Fakhimahmadi A, Nazmi F, Rahmati M, Bonab NM, Hashemi M, Moosavi MA. Nucleostemin silencing induces differentiation and potentiates all-trans-retinoic acid effects in human acute promyelocytic leukemia NB4 cells via autophagy. Leuk Res 2017; 63:15-21. [PMID: 29096331 DOI: 10.1016/j.leukres.2017.10.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 10/19/2017] [Accepted: 10/24/2017] [Indexed: 12/15/2022]
Abstract
Here, we report that targeting Nucleostemin (NS), a recently discovered stem cells-enriched gene, by a specific small interference RNA (siNS), decreases the rate of proliferation of acute promyelocytic leukemia (APL) NB4 cells and induces differentiation and autophagy. In addition, NS silencing promotes the effects of all-trans-retinoic acid (ATRA)-based differentiation therapy in NB4 cells. Autophagy inhibitors 3-methyladenine and bafilomycin block the effect of NS targeting on differentiation, indicating a new functional link between NS and autophagy as an important regulator of differentiation in NB4 cells. The capability of NS in modulating autophagy and differentiation, alone or in combination with ATRA, may help to broaden the range of treatment options available to treat leukemia.
Collapse
Affiliation(s)
- Aila Fakhimahmadi
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, P.O. Box:14965/161, Tehran, Iran; Islamic Azad University Tehran Medical Branch, Tehran, Iran
| | - Farinaz Nazmi
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, P.O. Box:14965/161, Tehran, Iran; Department of Biology, Faculty of Natural Science, University of Tabriz, P.O. Box 51666-16471, Tabriz, Iran
| | - Marveh Rahmati
- Cancer Biology Research Center, Tehran University of Medical Sciences, Tehran, Iran; Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Moghtaran Bonab
- Department of Biology, Faculty of Natural Science, University of Tabriz, P.O. Box 51666-16471, Tabriz, Iran
| | | | - Mohammad Amin Moosavi
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, P.O. Box:14965/161, Tehran, Iran.
| |
Collapse
|
24
|
Tang X, Zha L, Li H, Liao G, Huang Z, Peng X, Wang Z. Upregulation of GNL3 expression promotes colon cancer cell proliferation, migration, invasion and epithelial-mesenchymal transition via the Wnt/β-catenin signaling pathway. Oncol Rep 2017; 38:2023-2032. [PMID: 28849076 PMCID: PMC5652940 DOI: 10.3892/or.2017.5923] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/03/2017] [Indexed: 11/09/2022] Open
Abstract
G protein nucleolar 3 (GNL3), a nucleolar GTP-binding protein, is highly expressed in progenitor cells, stem cells, and various types of cancer cells. Therefore, it is considered to have an important role in cancer pathogenesis. GNL3 has been reported to play crucial roles in cell proliferation, cell cycle regulation, inhibition of differentiation, ribosome biogenesis, and the maintenance of stemness, genome stability and telomere integrity. Furthermore, GNL3 has recently been shown to be involved in cancer invasion and metastasis. However, the biological significance of GNL3 in the invasion and metastasis of colon cancer remains unclear. This study was performed to address this gap in knowledge. GNL3 expression was upregulated in colon cancer tissue specimens and correlated with tumor differentiation, invasion and metastasis. GNL3 overexpression promoted cell proliferation, invasion, migration and the epithelial-mesenchymal transition (EMT) in colon cancer cells. Moreover, inhibition of the EMT and the Wnt/β-catenin signaling pathway induced by GNL3 knockdown was partially reversed by lithium chloride (LiCl). Based on these data, GNL3 promotes the EMT in colon cancer by activating the Wnt/β-catenin signaling pathway. In summary, GNL3 is upregulated in colon cancer and plays an important role in tumor growth, invasion and metastasis. Strategies targeting GNL3 are potential treatments for colon cancer.
Collapse
Affiliation(s)
- Xi Tang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Lang Zha
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hui Li
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Gang Liao
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Zhen Huang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xudong Peng
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Ziwei Wang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
25
|
Onderak AM, Anderson JT. Loss of the RNA helicase SKIV2L2 impairs mitotic progression and replication-dependent histone mRNA turnover in murine cell lines. RNA (NEW YORK, N.Y.) 2017; 23:910-926. [PMID: 28351885 PMCID: PMC5435864 DOI: 10.1261/rna.060640.117] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 03/02/2017] [Indexed: 06/06/2023]
Abstract
RNA surveillance via the nuclear exosome requires cofactors such as the helicase SKIV2L2 to process and degrade certain noncoding RNAs. This research aimed to characterize the phenotype associated with RNAi knockdown of Skiv2l2 in two murine cancer cell lines: Neuro2A and P19. SKIV2L2 depletion in Neuro2A and P19 cells induced changes in gene expression indicative of cell differentiation and reduced cellular proliferation by 30%. Propidium iodide-based cell-cycle analysis of Skiv2l2 knockdown cells revealed defective progression through the G2/M phase and an accumulation of mitotic cells, suggesting SKIV2L2 contributes to mitotic progression. Since SKIV2L2 targets RNAs to the nuclear exosome for processing and degradation, we identified RNA targets elevated in cells depleted of SKIV2L2 that could account for the observed twofold increase in mitotic cells. Skiv2l2 knockdown cells accumulated replication-dependent histone mRNAs, among other RNAs, that could impede mitotic progression and indirectly trigger differentiation.
Collapse
Affiliation(s)
- Alexis M Onderak
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin 53201, USA
| | - James T Anderson
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin 53201, USA
| |
Collapse
|
26
|
Hua L, Hu B, Yan D, Liu J, Shen Y, Zhao F, Shen C, Chen B, Cui X. Upregulated expression of Nucleostemin/GNL3 is associated with poor prognosis and Sorafenib Resistance in Hepatocellular Carcinoma. Pathol Res Pract 2017; 213:688-697. [PMID: 28476378 DOI: 10.1016/j.prp.2016.11.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 11/06/2016] [Accepted: 11/11/2016] [Indexed: 12/13/2022]
Abstract
Nucleostemin (NS)/GNL3 protein has been recently documented to be a nucleolar protein that was abundantly expressed in stem cells and cancer cells. Herein, we showed that NS was upregulated in HCC tissues and the expression of NS was inversely correlated with that of p53. Overexpression of NS predicted significantly worsened prognosis in HCC patients, suggesting that NS might serve as a prognostic marker of HCC. In addition, we found that depletion of NS sensitized HCC cells to sorafenib-induced apoptosis. Moreover, we found that the mechanism underlying NS-mediated sorafenib resistance involved dysregulated expression of p53, and downstream Bax and Bcl-2 proteins. NS interacted with p53 in HCC cells. Depletion of NS increased the expression of p53 and Bax, whereas impaired the level of cellular Bcl-2. Interference of NS enhanced the cytotoxic effects of sorafenib in HCC cells. Furthermore, ectopic expression of NS impaired the apoptosis of HCC cells following sorafenib exposure. Therefore, NS may contribute to sorafenib resistance in HCC cells through the modulation of p53 pathway and Bcl-2 proteins. These findings indicated that the combination of silencing NS expression and sorafenib treatment is a promising therapeutic strategy in treatment of HCC.
Collapse
Affiliation(s)
- Lu Hua
- Department of Radiotherapy, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, Jiangsu Province, China
| | - Baoying Hu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, Jiangsu Province, China; Basic Medical Research Centre, Medical College, Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Daliang Yan
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, Jiangsu Province, China
| | - Jinxia Liu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, Jiangsu Province, China
| | - Yifen Shen
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, Jiangsu Province, China
| | - Fengbo Zhao
- Basic Medical Research Centre, Medical College, Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Chaoyan Shen
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, Jiangsu Province, China
| | - Buyou Chen
- Department of Radiotherapy, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China.
| | - Xiaopeng Cui
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China.
| |
Collapse
|
27
|
Zhao S, Xia Y, Zhang F, Xiong Z, Li Y, Yan W, Chen X, Wang W, Wang H, Gao E, Lee Y, Li C, Wang S, Zhang L, Tao L. Nucleostemin dysregulation contributes to ischemic vulnerability of diabetic hearts: Role of ribosomal biogenesis. J Mol Cell Cardiol 2017; 108:106-113. [PMID: 28549781 DOI: 10.1016/j.yjmcc.2017.05.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/29/2017] [Accepted: 05/22/2017] [Indexed: 01/20/2023]
Abstract
Diabetes is a major health problem worldwide. As well-known, diabetes greatly increases cardiac vulnerability to ischemia/reperfusion (I/R) injury, but the underlying mechanisms remain elusive. Nucleostemin (NS) is a nucleolar protein that controls ribosomal biogenesis and exerts cardioprotective effects against I/R injury. However, whether NS-mediated ribosomal biogenesis regulates ischemic vulnerability of diabetic hearts remains unanswered. Utilizing myocardial I/R mouse models, we found that cardiac NS expression significantly increased in response to I/R in normal diet (ND)-fed mice. Surprisingly, cardiac NS failed to be upregulated in high fat diet (HFD)-induced diabetic mice, accompanied by obvious ribosomal dysfunction. Compared with ND group, cardiac specific overexpression of NS by adenovirus (AV) injection significantly restored I/R-induced ribosomal function enhancement, reduced cardiomyocyte apoptosis, improved cardiac function, and decreased infarct sizes in diabetic mice. Notably, co-treatment of homoharringtonine (HHT), a selective inhibitor of ribosomal function, totally blocked NS-mediated cardioprotective effects against I/R injury. Furthermore, in cultured cardiomyocytes, saturated fatty acids treatment, but not high glucose exposure, significantly inhibited simulated I/R-induced NS upregulation and ribosomal function improvement. In conclusion, these data for the first time demonstrate that NS dysregulation induced by saturated fatty acids exposure might be an important cause of increased ischemic vulnerability to I/R injury in diabetic hearts. Targeting NS dysregulation and subsequent ribosomal dysfunction could be a promising therapeutic strategy for diabetic I/R injury management.
Collapse
Affiliation(s)
- Shihao Zhao
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Yunlong Xia
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Fuyang Zhang
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China; Department of Physiology, the Fourth Military Medical University, China; Department of Cardiology, the 201st Hospital of People's Liberation Army, China
| | - Zhenyu Xiong
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Yueyang Li
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Wenjun Yan
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Xiyao Chen
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, China
| | - Wei Wang
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Helin Wang
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Erhe Gao
- Center for Translational Medicine, Temple University, United States
| | - Yan Lee
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Shan Wang
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Ling Zhang
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China.
| |
Collapse
|
28
|
Hu B, Hua L, Ni W, Wu M, Yan D, Chen Y, Lu C, Chen B, Wan C. Nucleostemin/GNL3 promotes nucleolar polyubiquitylation of p27 kip1 to drive hepatocellular carcinoma progression. Cancer Lett 2016; 388:220-229. [PMID: 27998760 DOI: 10.1016/j.canlet.2016.12.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 11/09/2016] [Accepted: 12/09/2016] [Indexed: 01/31/2023]
Abstract
p27kip, as a cyclin dependent kinase inhibitor (CDKI), plays a pivotal role in the regulation of cell cycle progression and hepatocarcinogenesis. Herein, we revealed that p27 exhibited apparent nucleolar distribution and interacted with nucleolar protein nucleostemin (NS) in Hepatocellular carcinoma (HCC) cells. Furthermore, subcellular fractionation experiments demonstrated that nucleolar p27 had significantly higher level of polyubiquitylation, compared with nucleoplasmic fraction. Depletion of NS inhibited nucleolar polyubiquitylation of p27, indicating an involvement of NS in triggering p27 ubiquitylation and inactivation during HCC development. Moreover, we found that knockdown of NS promoted p27 to bind to CDK2-Cyclin E complex and inhibited the activity of CDK2, resulting in consequent cell cycle arrest in HCC cells. Furthermore, silencing NS expression reduced in vitro colony formation and in vivo tumor growth of HCC cells. Finally, we found that NS was upregulated in HCC tissues, compared with adjacent non-tumorous tissues. Kaplan-Meier analysis indicated patients with high expression of NS and low expression of p27 had significantly worsened prognosis. Our results suggested NS mediated p27-dependent cell cycle control via inducing nucleolar sequestration and polyubiquitylation of p27 in HCC. These findings help gain an insightful view into the mechanism underlying aberrant cell cycle progression during hepatocarcinogenesis, and thus benefit the development of molecular-targeted therapies in HCC.
Collapse
Affiliation(s)
- Baoying Hu
- Department of Radiotherapy, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, Jiangsu Province, China; Basic Medical Research Centre, Medical College, Nantong University, Nantong 226001, Jiangsu Province, China
| | - Lu Hua
- Department of Radiotherapy, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, Jiangsu Province, China
| | - Wenkai Ni
- Department of Gastroenterlogy, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Miaomiao Wu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, Jiangsu Province, China
| | - Daliang Yan
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, Jiangsu Province, China
| | - Yuyan Chen
- Class 2 Grade 13, Clinical Medicine, Medical College, Nantong University, Nantong 226001, Jiangsu Province, China
| | - Cuihua Lu
- Department of Gastroenterlogy, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Buyou Chen
- Department of Radiotherapy, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China.
| | - Chunhua Wan
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, Jiangsu Province, China.
| |
Collapse
|
29
|
Wang Y, DiMario P. Loss of Drosophila nucleostemin 2 (NS2) blocks nucleolar release of the 60S subunit leading to ribosome stress. Chromosoma 2016; 126:375-388. [PMID: 27150106 DOI: 10.1007/s00412-016-0597-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 04/18/2016] [Accepted: 04/25/2016] [Indexed: 12/24/2022]
Abstract
Four nucleostemin-like proteins (nucleostemin (NS) 1-4) were identified previously in Drosophila melanogaster. NS1 and NS2 are nucleolar proteins, while NS3 and NS4 are cytoplasmic proteins. We showed earlier that NS1 (homologous to human GNL3) enriches within the granular components (GCs) of Drosophila nucleoli and is required for efficient maturation or nucleolar release of the 60S subunit. Here, we show that NS2 is homologous to the human nucleostemin-like protein, Ngp1 (GNL2), and that endogenous NS2 is expressed in both progenitor and terminally differentiated cell types. Exogenous GFP-NS2 enriched within nucleolar GCs versus endogenous fibrillarin that marked the dense fibrillar components (DFCs). Like NS1, depletion of NS2 in midgut cells blocked the release of the 60S subunit as detected by the accumulation of GFP-RpL11 within nucleoli, and this likely led to the general loss of 60S subunits as shown by immunoblot analyses of RpL23a and RpL34. At the ultrastructural level, nucleoli in midgut cells depleted of NS2 displayed enlarged GCs not only on the nucleolar periphery but interspersed within the DFCs. Depletion of NS2 caused ribosome stress: larval midgut cells displayed prominent autophagy marked by the appearance of autolysosomes containing mCherry-ATG8a and the appearance of rough endoplasmic reticulum (rER)-derived isolation membranes. Larval imaginal wing disc cells depleted of NS2 induced apoptosis as marked by anti-caspase 3 labeling; loss of these progenitor cells resulted in defective adult wings. We conclude that nucleolar proteins NS1 and NS2 have similar but non-overlapping roles in the final maturation or nucleolar release of 60S ribosomal subunits.
Collapse
Affiliation(s)
- Yubo Wang
- Department of Biological Sciences, Louisiana State University, 202 Life Sciences Building, Baton Rouge, LA, 70803-1715, USA
| | - Patrick DiMario
- Department of Biological Sciences, Louisiana State University, 202 Life Sciences Building, Baton Rouge, LA, 70803-1715, USA.
| |
Collapse
|
30
|
Tsai RYL. Balancing self-renewal against genome preservation in stem cells: How do they manage to have the cake and eat it too? Cell Mol Life Sci 2016; 73:1803-23. [PMID: 26886024 PMCID: PMC5040593 DOI: 10.1007/s00018-016-2152-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 01/18/2016] [Accepted: 01/28/2016] [Indexed: 01/22/2023]
Abstract
Stem cells are endowed with the awesome power of self-renewal and multi-lineage differentiation that allows them to be major contributors to tissue homeostasis. Owing to their longevity and self-renewal capacity, they are also faced with a higher risk of genomic damage compared to differentiated cells. Damage on the genome, if not prevented or repaired properly, will threaten the survival of stem cells and culminate in organ failure, premature aging, or cancer formation. It is therefore of paramount importance that stem cells remain genomically stable throughout life. Given their unique biological and functional requirement, stem cells are thought to manage genotoxic stress somewhat differently from non-stem cells. The focus of this article is to review the current knowledge on how stem cells escape the barrage of oxidative and replicative DNA damage to stay in self-renewal. A clear statement on this subject should help us better understand tissue regeneration, aging, and cancer.
Collapse
Affiliation(s)
- Robert Y L Tsai
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University Health Science Center, 2121 W. Holcombe Blvd, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, 77843, USA.
| |
Collapse
|
31
|
Kodiha M, Mahboubi H, Maysinger D, Stochaj U. Gold Nanoparticles Impinge on Nucleoli and the Stress Response in MCF7 Breast Cancer Cells. Nanobiomedicine (Rij) 2016; 3:3. [PMID: 29942378 PMCID: PMC5998265 DOI: 10.5772/62337] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 02/01/2016] [Indexed: 01/15/2023] Open
Abstract
Cancer cells can take up gold nanoparticles of different morphologies. These particles interact with the plasma membrane and often travel to intracellular organelles. Among organelles, the nucleus is especially susceptible to the damage that is inflicted by gold nanoparticles. Located inside the nucleus, nucleoli are specialized compartments that transcribe ribosomal RNA genes, produce ribosomes and function as cellular stress sensors. Nucleoli are particularly prone to gold nanoparticle-induced injury. As such, small spherical gold nanoparticles and gold nanoflowers interfere with the transcription of ribosomal DNA. However, the underlying mechanisms are not fully understood. In this study, we examined the effects of gold nanoparticles on nucleolar proteins that are critical to ribosome biogenesis and other cellular functions. We show that B23/nucleophosmin, a nucleolar protein that is tightly linked to cancer, is significantly affected by gold nanoparticles. Furthermore, gold nanoparticles impinge on the cellular stress response, as they reduce the abundance of the molecular chaperone hsp70 and O-GlcNAc modified proteins in the nucleus and nucleoli. Together, our studies set the stage for the development of nanomedicines that target the nucleolus to eradicate proliferating cancer cells.
Collapse
Affiliation(s)
- Mohamed Kodiha
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Hicham Mahboubi
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Dusica Maysinger
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada
| | - Ursula Stochaj
- Department of Physiology, McGill University, Montreal, QC, Canada
| |
Collapse
|
32
|
Affiliation(s)
- R Y L Tsai
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, USA
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, USA
| |
Collapse
|
33
|
Abstract
Nucleostemin (NS) protects the genome from replication-induced DNA damage and has an indispensable role in maintaining the continuous proliferation of both p53-wild-type and mutant cells. Yet, some outcomes of NS-deficient cells appear to be shaped by their p53 status, which stimulates conflicting claims on the role of p53 in executing the NS function. This disparity was conveniently attributed to the usual suspect of cell-type variations. To provide a definitive resolution, we investigated the interplay between NS and p53 in two pairs of isogenic cells, that is, genetically modified mouse embryonic fibroblast (MEF) cells and HCT116 human colon cancer cells. In MEF cells, p53 deletion further compromises rather than rescues the proliferative potential of NS-depleted cells without changing their G2/M arrest fate before prophase entry. The detrimental effect of p53 loss in NS-depleted MEF cells correlates with a dramatic increase of polyploid giant cells (PGCs) (up to 24%), which indicates aberrant mitosis. To determine how p53 shapes the response of cells to NS depletion at the molecular level, we showed that p53 turns on the expression of reprimo and MDM2 in NS-deficient MEF cells. In absence of p53, NS-deficient MEF cells exhibit increased levels of phosphorylated cdc2 (Y15) protein and cyclin B1. In cancer (HCT116) cells, NS loss leads to G2/M arrest under both p53wt and p53ko conditions and increases phosphorylated cdc2 more in p53ko than in p53wt cells, as it does in MEF cells. Unlike its effect in MEF cells, NS depletion decreases tumor growth and increases the expression of reprimo and cyclin B1 in a p53-independent manner in HCT116 cells. Our data indicate that the p53 status of NS-deficient cells orchestrates how they respond to G2/M arrest in a normal versus cancer cell distinct fashion.
Collapse
Affiliation(s)
- Guanqun Huang
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA; Department of general surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510700, China
| | - Lingjun Meng
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA
| | - Robert Y L Tsai
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA; Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, TX 77054, USA
| |
Collapse
|
34
|
Nazmi F, Moosavi MA, Rahmati M, Hoessinpour-Feizi MA. Modeling and structural analysis of human Guanine nucleotide-binding protein-like 3,nucleostemin. Bioinformation 2015; 11:353-8. [PMID: 26339152 PMCID: PMC4546995 DOI: 10.6026/97320630011353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 06/28/2015] [Accepted: 06/29/2015] [Indexed: 11/25/2022] Open
Abstract
Human GNL3 (nucleostemin) is a recently discovered nucleolar protein with pivotal functions in maintaining genomic integrity
and determining cell fates of various normal and cancerous stem cells. Recent reports suggest that targeting this GTP-binding
protein may have therapeutic value in cancer. Although, sequence analyzing revealed that nucleostemin (NS) comprises 5
permuted GTP-binding motifs, a crystal structure for this protein is missing at Protein Data Bank (PDB). Obviously, any attempt
for predicting of NS structure can further our knowledge on its functional sites and subsequently designing molecular inhibitors.
Herein, we used bioinformatics tools and could model 262 amino acids of NS (132-393 aa). Initial models were built by
MODELLER, refined with Scwrl4 program, and validated with ProsA and Jcsc databases as well as PSVS software. Then, the best
quality model was chosen for motif and domain analyzing by Pfam, PROSITE and PRINTS. The final model was visualized by
vmd program. This predicted model may pave the way for next studies regarding ligand binding states and interaction sites as
well as screening of databases for potential inhibitors.
Collapse
Affiliation(s)
- Farinaz Nazmi
- Department of Molecular Medicine, Institute of Medical biotechnology, National Institute for genetic Engineering and biotechnology, Tehran, Iran ; Department of Zoology, Faculty of Natural Science, The University of Tabriz, Iran
| | - Mohammad Amin Moosavi
- Department of Molecular Medicine, Institute of Medical biotechnology, National Institute for genetic Engineering and biotechnology, Tehran, Iran ; Hematology and Oncology Research Center, Tabriz University of medical Science, Tabriz, Iran
| | - Marveh Rahmati
- Hematology and Oncology Research Center, Tabriz University of medical Science, Tabriz, Iran
| | | |
Collapse
|
35
|
Tsai RYL. Pluripotency Versus Self-Renewal of ES Cells: Two Sides of the Same Coin or More? Stem Cells 2015; 33:2358-9. [PMID: 25809666 DOI: 10.1002/stem.2008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 02/28/2015] [Indexed: 12/22/2022]
Affiliation(s)
- Robert Y L Tsai
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas, 77030, USA.,Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas, 77843, USA
| |
Collapse
|
36
|
Tancioni I, Miller NLG, Uryu S, Lawson C, Jean C, Chen XL, Kleinschmidt EG, Schlaepfer DD. FAK activity protects nucleostemin in facilitating breast cancer spheroid and tumor growth. Breast Cancer Res 2015; 17:47. [PMID: 25880415 PMCID: PMC4407832 DOI: 10.1186/s13058-015-0551-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 03/11/2015] [Indexed: 12/17/2022] Open
Abstract
Introduction Focal adhesion kinase (FAK) controls cell growth and survival downstream of integrin-matrix receptors. Upon adhesion loss or FAK inhibition, FAK can translocate to the nucleus. The nucleolus is a non-membrane nuclear structure that regulates ribosome biogenesis and cell proliferation. Nucleostemin (NS), a nucleolar-localized protein, modulates cell cycle progression, stemness, and three-dimensional tumor spheroid formation. The signaling pathways that regulate NS levels in tumors remain undefined. Methods Human breast carcinoma cells were evaluated for growth in culture (adherent and anchorage-independent spheroid) and as orthotopic tumors. FAK signaling was evaluated by pharmacological FAK inhibitor addition (PF-271, IC50 ~ 0.1 μM) and by small hairpin RNA (shRNA) knockdown followed by re-expression of FAK wildtype (WT) or a kinase-dead (KD, K454R) FAK point mutant. Immunoblotting was used to evaluate FAK, NS, nucleolar phosphoprotein B23, and nucleolin levels. Total and phosphospecific antibody imunoblotting were used to detect changes in FAK, Akt kinase (Akt also known as protein kinase B), and 4E-binding protein 1 (4E-BP1) phosphorylation, a translation repressor protein and target of the mammalian target of rapamycin (mTOR) complex. Immunohistochemical, co-immunoprecipitation, and cellular fractionation analyses were used to evaluate FAK association with nucleoli. Results Pharmacological (0.1 μM PF-271) or genetic inhibition of FAK activity prevents MDA-MB-231 and 4T1L breast carcinoma growth as spheroids and as orthotopic tumors. FAK inhibition triggers proteasome-mediated decreased NS levels but no changes in other nucleolar proteins such as B23 (nucleophosmin) or nucleolin. Active FAK was associated with purified nucleoli of anchorage-independent cells and present within nucleoli of human invasive ductal carcinoma tumor samples. FAK co-immunoprecipitated with B23 that binds NS and a complex between FAK, NS, Akt, and mTOR was detected. Constitutively-active Akt kinase promoted tumor spheroid growth, stabilized NS levels, and promoted pS65 4E-BP1 phosphorylation in the presence of inhibited FAK. Rapamycin lowered NS levels and inhibited pS65 4E-BP1 phosphorylation in cells with activated Akt-mTOR signaling. Conclusions FAK signaling occurs in the nucleolus, active FAK protects NS, and Akt-mTOR pathway regulates NS protein stability needed for breast carcinoma spheroid and tumor growth. Electronic supplementary material The online version of this article (doi:10.1186/s13058-015-0551-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Isabelle Tancioni
- Department of Reproductive Medicine, University of California San Diego, Moores Cancer Center, 3855 Health Sciences Dr, La Jolla, CA, 92093, USA.
| | - Nichol L G Miller
- Department of Reproductive Medicine, University of California San Diego, Moores Cancer Center, 3855 Health Sciences Dr, La Jolla, CA, 92093, USA. .,Current address: Pfizer, La Jolla, CA, 92121, USA.
| | - Sean Uryu
- Department of Reproductive Medicine, University of California San Diego, Moores Cancer Center, 3855 Health Sciences Dr, La Jolla, CA, 92093, USA.
| | - Christine Lawson
- Department of Reproductive Medicine, University of California San Diego, Moores Cancer Center, 3855 Health Sciences Dr, La Jolla, CA, 92093, USA.
| | - Christine Jean
- Department of Reproductive Medicine, University of California San Diego, Moores Cancer Center, 3855 Health Sciences Dr, La Jolla, CA, 92093, USA. .,Current address: INSERM U1037 - Cancer Research Center, Toulouse, France.
| | - Xiao Lei Chen
- Department of Reproductive Medicine, University of California San Diego, Moores Cancer Center, 3855 Health Sciences Dr, La Jolla, CA, 92093, USA.
| | - Elizabeth G Kleinschmidt
- Department of Reproductive Medicine, University of California San Diego, Moores Cancer Center, 3855 Health Sciences Dr, La Jolla, CA, 92093, USA.
| | - David D Schlaepfer
- Department of Reproductive Medicine, University of California San Diego, Moores Cancer Center, 3855 Health Sciences Dr, La Jolla, CA, 92093, USA.
| |
Collapse
|