1
|
Chen H, Zhang L, Yue F, Cui C, Li Y, Zhang Q, Liang L, Meng L, Zhang C. Effects of assisted reproductive technology on gene expression in heart and spleen tissues of adult offspring mouse. Front Endocrinol (Lausanne) 2023; 14:1035161. [PMID: 37065763 PMCID: PMC10098333 DOI: 10.3389/fendo.2023.1035161] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 03/10/2023] [Indexed: 04/18/2023] Open
Abstract
OBJECTIVES Assisted reproductive technology (ART) is an important part of reproductive medicine, whose possible effects on offspring's health have drawn widespread attention in recent years. However, relevant studies are limited to postnatal short-term follow-up and lack of diverse sample sources analysis other than blood. METHODS In this study, a mouse model was used to explore the effects of ART on fetal development and gene expression in the organs of offspring in the adulthood using next-generation sequencing. The sequencing results were then analyzed. RESULTS The results showed that it caused abnormal expression in 1060 genes and 179 genes in the heart and spleen, respectively. Differentially expressed genes (DEGs) in the heart are mainly enriched in RNA synthesis and processing, and the cardiovascular system development also shows enrichment. STRING analysis identified Ccl2, Ptgs2, Rock1, Mapk14, Agt, and Wnt5a as the core interacting factors. DEGs in the spleen are significantly enriched in anti-infection and immune responses, which include the core factors Fos, Jun and Il1r2. Further exploration revealed the abnormal expression of 42 and 5 epigenetic modifiers in the heart and spleen, respectively. The expression of the imprinted genes Dhcr7, Igf2, Mest and Smoc1 decreased in the hearts of ART offspring, and the DNA methylation levels of Igf2- and Mest-imprinting control regions (ICRs) increased abnormally. CONCLUSION In the mouse model, ART can interfere with the gene expression pattern in the heart and spleen of the adult offspring and that these changes are related to the aberrant expression of epigenetic regulators.
Collapse
Affiliation(s)
- Huanhuan Chen
- Reproductive Medicine Center, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan, China
| | - Lei Zhang
- Reproductive Medicine Center, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan, China
| | - Feng Yue
- Reproductive Medicine Center, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan, China
| | - Chenchen Cui
- Reproductive Medicine Center, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan, China
| | - Yan Li
- Reproductive Medicine Center, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan, China
| | - Qingwen Zhang
- Reproductive Medicine Center, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan, China
| | - Linlin Liang
- Reproductive Medicine Center, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan, China
| | - Li Meng
- Reproductive Medicine Center, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan, China
- *Correspondence: Li Meng, ; Cuilian Zhang,
| | - Cuilian Zhang
- Reproductive Medicine Center, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan, China
- *Correspondence: Li Meng, ; Cuilian Zhang,
| |
Collapse
|
2
|
Kadota Y, Kawakami T, Sato M, Suzuki S. Mouse mesoderm-specific transcript inhibits adipogenic differentiation and induces trans-differentiation into hepatocyte-like cells in 3T3-L1 preadiocytes. BMC Res Notes 2022; 15:164. [PMID: 35538505 PMCID: PMC9092885 DOI: 10.1186/s13104-022-06051-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/26/2022] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE The mesoderm-specific transcript (Mest) is an imprinted gene that is transcribed from the paternal allele. It is a marker of adipose tissue expansion; however, it is uncertain whether Mest expression promotes or suppresses adipogenic differentiation. To elucidate the effects of Mest expression on adipogenic differentiation, we transfected an expression vector or siRNA for mouse Mest into 3T3-L1 mouse preadipocyte cell line. RESULTS In differentiated 3T3-L1 adipocytes, Mest overexpression decreased lipid accumulation. Conversely, gene silencing of Mest increased the accumulation of lipid droplets in adipocytes. These results demonstrate that Mest negatively regulates adipocyte differentiation. Further, Mest induced trans-differentiation of 3T3-L1 cells into hepatocytes, and its overexpression induced the expression of hepatocyte marker genes, including albumin and α-fetoprotein. In the presence of dexamethasone, the forced expression of the Mest caused morphological changes in 3T3-L1 cells. Cells were flat and polygonal shapes, with an increased accumulation of intracellular glycogen and other features that are typical of hepatocytes. Therefore, Mest inhibits adipogenic differentiation of 3T3-L1 preadipocytes by inducing hepatocyte trans-differentiation.
Collapse
Affiliation(s)
- Yoshito Kadota
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Yamashiro-cho, Tokushima, Japan
| | - Takashige Kawakami
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Yamashiro-cho, Tokushima, Japan
| | - Masao Sato
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Yamashiro-cho, Tokushima, Japan
| | - Shinya Suzuki
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Yamashiro-cho, Tokushima, Japan
| |
Collapse
|
3
|
Velker BAM, Denomme MM, Krafty RT, Mann MRW. Maintenance of Mest imprinted methylation in blastocyst-stage mouse embryos is less stable than other imprinted loci following superovulation or embryo culture. ENVIRONMENTAL EPIGENETICS 2017; 3:dvx015. [PMID: 29492315 PMCID: PMC5804554 DOI: 10.1093/eep/dvx015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 06/07/2017] [Accepted: 07/19/2017] [Indexed: 06/08/2023]
Abstract
Assisted reproductive technologies are fertility treatments used by subfertile couples to conceive their biological child. Although generally considered safe, these pregnancies have been linked to genomic imprinting disorders, including Beckwith-Wiedemann and Silver-Russell Syndromes. Silver-Russell Syndrome is a growth disorder characterized by pre- and post-natal growth retardation. The Mest imprinted domain is one candidate region on chromosome 7 implicated in Silver-Russell Syndrome. We have previously shown that maintenance of imprinted methylation was disrupted by superovulation or embryo culture during pre-implantation mouse development. For superovulation, this disruption did not originate in oogenesis as a methylation acquisition defect. However, in comparison to other genes, Mest exhibits late methylation acquisition kinetics, possibly making Mest more vulnerable to perturbation by environmental insult. In this study, we present a comprehensive evaluation of the effects of superovulation and in vitro culture on genomic imprinting at the Mest gene. Superovulation resulted in disruption of imprinted methylation at the maternal Mest allele in blastocysts with an equal frequency of embryos having methylation errors following low or high hormone treatment. This disruption was not due to a failure of imprinted methylation acquisition at Mest in oocytes. For cultured embryos, both the Fast and Slow culture groups experienced a significant loss of maternal Mest methylation compared to in vivo-derived controls. This loss of methylation was independent of development rates in culture. These results indicate that Mest is more susceptible to imprinted methylation maintenance errors compared to other imprinted genes.
Collapse
Affiliation(s)
- Brenna A. M. Velker
- Department of Obstetrics & Gynecology, University of Western Ontario, Schulich School of Medicine and Dentistry, London, ON, Canada
- Department of Biochemistry, University of Western Ontario, Schulich School of Medicine and Dentistry, London, ON, Canada
- Children’s Health Research Institute, London, ON, Canada
| | - Michelle M. Denomme
- Department of Obstetrics & Gynecology, University of Western Ontario, Schulich School of Medicine and Dentistry, London, ON, Canada
- Department of Biochemistry, University of Western Ontario, Schulich School of Medicine and Dentistry, London, ON, Canada
- Children’s Health Research Institute, London, ON, Canada
- Fertility Laboratories Of Colorado, 10290 Ridgegate Circle, Lonetree, CO 80124 USA
| | - Robert T. Krafty
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mellissa R. W. Mann
- Magee-Womens Research Institute, Pittsburgh, PA, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Dhasarathy A, Roemmich JN, Claycombe KJ. Influence of maternal obesity, diet and exercise on epigenetic regulation of adipocytes. Mol Aspects Med 2016; 54:37-49. [PMID: 27825817 DOI: 10.1016/j.mam.2016.10.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 10/25/2016] [Accepted: 10/25/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Archana Dhasarathy
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - James N Roemmich
- USDA-ARS-PA, Grand Forks Human Nutrition Research Center, 2420 2nd Avenue North, Grand Forks, ND 58203, USA
| | - Kate J Claycombe
- USDA-ARS-PA, Grand Forks Human Nutrition Research Center, 2420 2nd Avenue North, Grand Forks, ND 58203, USA.
| |
Collapse
|
5
|
Peng W, Chen Y, Luo X, Shan N, Lan X, Olson D, Zhang H, Ding YB, Qi HB. DNA methylation-associated repression of MEST/PEG1 expression contributes to the invasion of extravillous trophoblast cells. Placenta 2016; 46:92-101. [PMID: 27697227 DOI: 10.1016/j.placenta.2016.08.093] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 08/21/2016] [Accepted: 08/30/2016] [Indexed: 12/31/2022]
Abstract
INTRODUCTION The invasion of extravillous cytotrophoblasts (EVTs) into the maternal uterine decidua and vasculature is critical for human placenta development and pregnancy maintenance. The imprinted gene MEST/PEG1 has been implicated in trophoblast development; however, the role of MEST in EVT invasion and the accompanying early pregnancy complications are not fully understood. METHODS Western blot, immunofluorescence and immunohistochemistry were used to detect MEST protein expression and localization by using antibodies recognize 2 reported isoforms. Specific small interference RNA (siRNA) targeting both of the MEST isoforms was applied to silence MEST expression in extravillous explants and HTR8/SVneo cells. Cell invasion and migration were assessed using the Matrigel invasion, Transwell migration assay and the xCELLigence system. Promoter DNA methylation was examined using bisulfite-sequencing polymerase chain reaction (BSP). RESULTS MEST protein was highly expressed in EVTs in the first trimester placenta and in the invasive EVT cell lines HTR-8/Svneo and HPT-8. Weak MEST expression was found in cytotrophoblasts (CTBs) and the choriocarcinoma-derived CTB cell line JEG-3. The specific siRNA knockdown of MEST expression significantly reduced HTR-8/Svneo cell invasion and migration as well as extravillous explant outgrowth, which were associated with the downregulation of Twist, N-cadherin and Vimentin. Decreased MEST protein expression with isoform 2 promoter hypermethylation was observed in the placentas of missed abortions, suggesting a possible pathological mechanism of missed abortion. CONCLUSIONS Suppressed expression of MEST was associated with its isoform 2 promoter hypermethylation ex vivo placenta tissues and in vitro cultured EVT cell lines. The present results provide a possible pathological mechanism of missed abortion.
Collapse
Affiliation(s)
- Wei Peng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China; Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Ying Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China; Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xin Luo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China; Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Nan Shan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China; Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xi Lan
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, China
| | - David Olson
- Departments of Obstetrics and Gynecology, Pediatrics and Physiology, University of Alberta, Edmonton, AB T6G 2S2, Canada; Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Hua Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China; Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yu-Bin Ding
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, China; Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China.
| | - Hong-Bo Qi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China; Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
6
|
Kadota Y, Yanagawa M, Nakaya T, Kawakami T, Sato M, Suzuki S. Gene expression of mesoderm-specific transcript is upregulated as preadipocytes differentiate to adipocytes in vitro. J Physiol Sci 2012; 62:403-11. [PMID: 22753118 PMCID: PMC10717349 DOI: 10.1007/s12576-012-0217-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 06/04/2012] [Indexed: 12/01/2022]
Abstract
Mesoderm-specific transcript (Mest) is a distinct gene associated with adipocyte differentiation and proliferation. The mechanisms regulating expression of the Mest gene are not established. Therefore, we investigated Mest gene expression during adipogenic differentiation in murine 3T3-L1 preadipocytes and adipose-derived stromal cells (ADCs) from C57BL/6J mouse adipose tissue. Expression of Mest mRNA increased significantly in 3T3-L1 cells during differentiation. Additionally, Mest mRNA expression levels were additively enhanced by the inhibition of DNA methylation. Expression levels of the Mest gene were also markedly elevated in differentiating ADCs in vitro. Additionally, we showed that Mest mRNA can be upregulated by increasing intracellular cAMP, and that Mest expression is suppressed by inhibition of protein kinase A (PKA). Mest expression was regulated through cAMP-dependent PKA pathways during differentiation of preadipocytes into adipocytes in vitro, supporting the critical role of Mest in proliferation and differentiation of adipocytes.
Collapse
Affiliation(s)
- Yoshito Kadota
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Yamashiro-cho, Tokushima, 770-8514, Japan.
| | | | | | | | | | | |
Collapse
|
7
|
[Studies on SNP and genomic imprinting of the PEG1 gene in swine]. YI CHUAN = HEREDITAS 2011; 33:738-42. [PMID: 22049687 DOI: 10.3724/sp.j.1005.2011.00738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PEG1 affects animal embryo growth and maternal behavior. The paternal allele-specific expression of PEG1 was reported in most animal species, but the expressive pattern of PEG1 was not clear in piglets born. In order to investigate the imprinting pattern of PEG1 in pig, 166 samples were used to SNP of PEG1 from Landrace, Yorkshire, and Lantang breeds by PCR-SSCP. Single nucleotide polymorphism (SNP) of PEG1 in exon 12 was identfied. The genotype frequency and the expressive pattern were anslyzed in swine by RT-PCR-RFLP/SSCP. One SNP (a G-->A transition) was identified in exon 12 of PEG1. Maternal expression of PEG1 exon 12 was observed in all major organs (stomach, thymus, pancreas, spleen, lung, muscle, liver, tongue, kidney, brain, bladder, and heart) and placenta of three heterozygous pigs. PEG1 was paternally expressed and maternally imprinted in swine.
Collapse
|
8
|
Koza RA, Rogers P, Kozak LP. Inter-individual variation of dietary fat-induced mesoderm specific transcript in adipose tissue within inbred mice is not caused by altered promoter methylation. Epigenetics 2010; 4:512-8. [PMID: 19875931 DOI: 10.4161/epi.4.7.10031] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Mesoderm specific transcript (Mest), an imprinted gene associated with fat mass expansion under conditions of positive energy balance, shows highly variable expression (approximately 80-fold) in white adipose tissue (WAT) of C57BL/6J (B6) mice fed an obesogenic diet. Since B6 mice are essentially genetically invariant and Mest is known to be regulated by CpG methylation within its immediate proximal promoter, the large variability in its expression in adipose tissue has the hallmarks of being controlled via an epigenetic mechanism. In this study, bisulfite sequencing and allelic discrimination analyses were performed to determine whether variations in CpG methylation within the Mest promoter were associated with its expression. Results showed no relationship between CpG methylation in the Mest promoter and high versus low expression in either WAT or isolated adipocytes; and, experiments using a single nucleotide polymorphism in the Mest promoter region between B6 and Castaneus mice showed the expected pattern for an imprinted gene with all maternal alleles being methylated. These data suggest that mechanisms independent of the CpG methylation status of the Mest promoter must underlie the control of its expression during adipose tissue expansion.
Collapse
Affiliation(s)
- Robert A Koza
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA.
| | | | | |
Collapse
|
9
|
Katari S, Turan N, Bibikova M, Erinle O, Chalian R, Foster M, Gaughan JP, Coutifaris C, Sapienza C. DNA methylation and gene expression differences in children conceived in vitro or in vivo. Hum Mol Genet 2009; 18:3769-78. [PMID: 19605411 PMCID: PMC2748887 DOI: 10.1093/hmg/ddp319] [Citation(s) in RCA: 304] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 06/18/2009] [Accepted: 07/09/2009] [Indexed: 12/21/2022] Open
Abstract
Epidemiological data indicate that children conceived in vitro have a greater relative risk of low birth-weight, major and minor birth defects, and rare disorders involving imprinted genes, suggesting that epigenetic changes may be associated with assisted reproduction. We examined DNA methylation at more than 700 genes (1536 CpG sites) in placenta and cord blood and measured gene expression levels of a subset of genes that differed in methylation levels between children conceived in vitro versus in vivo. Our results suggest that in vitro conception is associated with lower mean methylation at CpG sites in placenta and higher mean methylation at CpG sites in cord blood. We also find that in vitro conception-associated DNA methylation differences are associated with gene expression differences at both imprinted and non-imprinted genes. The range of inter-individual variation in gene expression of the in vitro and in vivo groups overlaps substantially but some individuals from the in vitro group differ from the in vivo group mean by more than two standard deviations. Several of the genes whose expression differs between the two groups have been implicated in chronic metabolic disorders, such as obesity and type II diabetes. These findings suggest that there may be epigenetic differences in the gametes or early embryos derived from couples undergoing treatment for infertility. Alternatively, assisted reproduction technology may have an effect on global patterns of DNA methylation and gene expression. In either case, these differences or changes may affect long-term patterns of gene expression.
Collapse
Affiliation(s)
- Sunita Katari
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Nahid Turan
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | - Oluwatoyin Erinle
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Raffi Chalian
- Department of Obstetrics & Gynecology, University of Pennsylvania School of Medicine, Philadelphia, Philadelphia, PA 19104, USA
| | - Michael Foster
- Department of Obstetrics & Gynecology, University of Pennsylvania School of Medicine, Philadelphia, Philadelphia, PA 19104, USA
| | - John P. Gaughan
- Biostatistics Consulting Center, Temple University School of Medicine, Philadelphia, PA, USA and
| | - Christos Coutifaris
- Department of Obstetrics & Gynecology, University of Pennsylvania School of Medicine, Philadelphia, Philadelphia, PA 19104, USA
| | - Carmen Sapienza
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
10
|
Regenbrecht CRA, Lehrach H, Adjaye J. Stemming cancer: functional genomics of cancer stem cells in solid tumors. ACTA ACUST UNITED AC 2009; 4:319-28. [PMID: 18561035 PMCID: PMC2758383 DOI: 10.1007/s12015-008-9034-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer stem cells (CSCs) were discovered about 15 years ago in hematopoietic cancers. Subsequently, cancer stem cells were discovered in various solid tumors. Based on parallels with normal stem cells, a developmental process of cancer stem cells follows paths of organized, hierarchical structure of cells with different degrees of maturity. While some investigators have reported particular markers as identification of cancer stem cells, these markers require further research. In this review, we focus on the functional genomics of cancer stem cells. Functional genomics provides useful information on the signaling pathways which are consecutively activated or inactivated amongst those cells. This information is of particular importance for cancer research and clinical treatment in many respects. (1) Understanding of self-renewal mechanisms crucial to tumor growth. (2) Allow the identification of new, more specific marker for CSCs, and (3) pathways that are suitable as future targets for anti-cancer drugs. This is of particular importance, because today's chemotherapy targets the proliferating cancer cells sparing the relatively slow dividing cancer stem cells. The first step on this long road therefore is to analyze genome-wide expression-profiles within the same type of cancer and then between different types of cancer, encircling those target genes and pathways, which are specific to these cells.
Collapse
Affiliation(s)
- C R A Regenbrecht
- Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics (Molecular Embryology and Aging group), Ihnestrasse 73, 14195 Berlin, Germany
| | | | | |
Collapse
|
11
|
Sathyan P, Golden HB, Miranda RC. Competing interactions between micro-RNAs determine neural progenitor survival and proliferation after ethanol exposure: evidence from an ex vivo model of the fetal cerebral cortical neuroepithelium. J Neurosci 2007; 27:8546-57. [PMID: 17687032 PMCID: PMC2915840 DOI: 10.1523/jneurosci.1269-07.2007] [Citation(s) in RCA: 218] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2006] [Revised: 06/17/2007] [Accepted: 06/26/2007] [Indexed: 12/28/2022] Open
Abstract
The fetal brain is sensitive to a variety of teratogens, including ethanol. We showed previously that ethanol induced mitosis and stem cell maturation, but not death, in fetal cerebral cortex-derived progenitors. We tested the hypothesis that micro-RNAs (miRNAs) could mediate the teratogenic effects of ethanol in a fetal mouse cerebral cortex-derived neurosphere culture model. Ethanol, at a level attained by alcoholics, significantly suppressed the expression of four miRNAs, miR-21, -335, -9, and -153, whereas a lower ethanol concentration, attainable during social drinking, induced miR-335 expression. A GABA(A) receptor-dependent mechanism mediated miR-21, but not miR-335 suppression, suggesting that divergent mechanisms regulate ethanol-sensitive miRNAs. Antisense-mediated suppression of miR-21 expression resulted in apoptosis, suggesting that miR-21 is an antiapoptotic factor. miR-335 knockdown promoted cell proliferation and prevented death induced by concurrently suppressing miR-21, indicating that miR-335 is a proapoptotic, antimitogenic factor whose actions are antagonistic to miR-21. Computational analyses identified two genes, Jagged-1, a Notch-receptor ligand, and embryonic-lethal abnormal vision, Drosophila-like 2 (ELAVL2), a brain-specific regulator of RNA stability, as presumptive targets of three of four ethanol-sensitive micro-RNAs. Combined knockdown of miR-335, -21, and -153 significantly increased Jagged-1 mRNA. Furthermore, ethanol induced both Jagged-1 and ELAVL2 mRNA. The collective suppression of micro-RNAs is consistent with ethanol induction of cell cycle and neuroepithelial maturation in the absence of apoptosis. These data identify a role for micro-RNAs as epigenetic intermediaries, which permit teratogens to shape complex, divergent developmental processes, and additionally demonstrate that coordinately regulated miRNAs exhibit both functional synergy and antagonism toward each other.
Collapse
Affiliation(s)
- Pratheesh Sathyan
- Department Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Texas 77843-1114
| | - Honey B. Golden
- Department Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Texas 77843-1114
| | - Rajesh C. Miranda
- Department Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Texas 77843-1114
| |
Collapse
|
12
|
Kamei Y, Suganami T, Kohda T, Ishino F, Yasuda K, Miura S, Ezaki O, Ogawa Y. Peg1/Mestin obese adipose tissue is expressed from the paternal allele in an isoform-specific manner. FEBS Lett 2006; 581:91-6. [PMID: 17182038 DOI: 10.1016/j.febslet.2006.12.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2006] [Accepted: 12/04/2006] [Indexed: 10/23/2022]
Abstract
Paternally expressed 1 (Peg1)/mesoderm specific transcript (Mest) is an imprinted gene, which is only transcribed from the paternal (father's) allele. In some human cancer tissues, an alternatively spliced variant of PEG1/MEST mRNA using a different promoter of a distinct first exon is expressed from both paternal and maternal alleles. We previously reported that Peg1/Mest expression was markedly up-regulated in obese adipose tissue in mice. Moreover, transgenic overexpression of Peg1/Mest in the adipose tissue resulted in the enlargement of adipocytes in size. Given the potential pathophysiologic relevance in obesity, we examined the nature of increased expression of Peg1/Mest in obese adipose tissue. In obese adipose tissue, expression of Peg1/Mest was increased, but not that of other imprinted genes tested. The transcription rate of Peg1/Mest was increased in obese adipose tissue. We found at least four isoforms of mouse Peg1/Mest generated by use of the alternative first exons. We also demonstrated that the abundantly expressed Peg1/Mest in obese adipose tissue retained monoallelic expression. This is the first report of monoallelic induction of Peg1/Mest in adult tissues.
Collapse
Affiliation(s)
- Yasutomi Kamei
- Department of Molecular Medicine and Metabolism, Medical Research Institute, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Tokyo, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Baqir S, Smith LC. Inhibitors of histone deacetylases and DNA methyltransferases alter imprinted gene regulation in embryonic stem cells. CLONING AND STEM CELLS 2006; 8:200-13. [PMID: 17009896 DOI: 10.1089/clo.2006.8.200] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Pluripotent embryonic stem cells are able to differentiate into a variety of cell types, thereby making them a valuable source for transplantation medicine. Recent studies have reported the use of pharmacological agents, namely 5-Aza-Cytidine (5AzaC) and Trichostatin A (TSA), to guide embryonic stem (ES) cells to differentiate into specific cellular lineages. However, those drugs are known to be potent inhibitors of DNA methyltransferases and/or histone deacetylases. Since both epigenetic mechanisms are involved in the expression of imprinted genes in fetal and adult somatic tissues, it is essential to investigate further the role of these agents in regulating imprinted gene expression in embryonic cells. Embryonic stem cells were exposed to 5AzaC and TSA and analyzed for transcript abundance of a number of imprinted and non-imprinted marker genes. Most imprinted gene transcripts increased following exposure to 5AzaC or TSA alone and responded in either an additive or synergistic manner when exposed to both drugs together. Interestingly, transcript levels of several imprinted genes remained high and in some cases, increased further after drug removal or even after passaging the cells, indicating a long lasting and retarded effect on gene expression. Together, our results suggest that DNA methylation and histone acetylation play jointly an important epigenetic role in governing imprinted gene expression in embryonic stem cells. Moreover, these results describe the sensitivity and irreversibility of embryonic stem cells to epigenetic modifiers, highlighting potential risks for their use in therapeutic applications.
Collapse
Affiliation(s)
- Senan Baqir
- Centre de Recherche en Reproduction Animale (CRRA), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada
| | | |
Collapse
|
14
|
Takahashi M, Kamei Y, Ezaki O. Mest/Peg1 imprinted gene enlarges adipocytes and is a marker of adipocyte size. Am J Physiol Endocrinol Metab 2005; 288:E117-24. [PMID: 15353408 DOI: 10.1152/ajpendo.00244.2004] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Obesity is a common and serious metabolic disorder in the developed world that is occasionally accompanied by type II diabetes, atherosclerosis, hypertension, and hyperlipidemia. We have found that mesoderm-specific transcript (Mest)/paternally expressed gene 1 (Peg1) gene expression was markedly enhanced in white adipose tissue of mice with diet-induced and genetically caused obesity/diabetes but not with streptozotocin-induced diabetes, which does not cause obesity. Administration of pioglitazone, a drug for type II diabetes and activator of peroxisome proliferator-activated receptor (PPAR)gamma, in obese db/db mice reduced the enhanced expression of Mest mRNA in adipose tissue, concomitant with an increase in body weight and a decrease in the size of adipose cells. Ectopic expression of Mest in 3T3-L1 cells caused increased gene expression of adipose markers such as PPARgamma, CCAAT/enhancer binding protein (C/EBP)alpha, and adipocyte fatty acid binding protein (aP)2. In transgenic mice overexpressing Mest in adipose tissue, enhanced expression of the adipose genes was observed. Moreover, adipocytes were markedly enlarged in the transgenic mice. Thus Mest appears to enlarge adipocytes and could be a novel marker of the size of adipocytes.
Collapse
Affiliation(s)
- Mayumi Takahashi
- Division of Clinical Nutrition, National Institute of Health and Nutrition, 1-23-1, Toyama, Shinjuku-ku, Tokyo 162-8636, Japan
| | | | | |
Collapse
|
15
|
Kanwar YS, Kumar A, Ota K, Lin S, Wada J, Chugh S, Wallner EI. Identification of developmentally regulated mesodermal-specific transcript in mouse embryonic metanephros. Am J Physiol Renal Physiol 2002; 282:F953-65. [PMID: 11934706 DOI: 10.1152/ajprenal.00200.2001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mesodermal-specific cDNA or transcript (MEST) was identified by suppression subtractive hybridization-PCR of cDNA isolated from embryonic day 13 vs. newborn mice kidneys. At day 13 of mouse gestation, a high expression of MEST, with a single approximately 2.7-kb transcript that was exclusively localized to the metanephric mesenchyme was observed. The MEST mRNA expression gradually decreased during the later stages and then abruptly decreased in the newborn kidneys and subsequent postnatal life, after which a very mild expression persisted in the glomerular mesangium. Regression in mRNA expression during embryonic renal development appears to be related to methylation of the MEST gene. Treatment of metanephroi, harvested at day 13 of gestation with MEST-specific antisense oligodeoxynucleotide resulted in a dose-dependent decrease in the size of the explants and the nephron population. This was associated with a selective decrease in MEST mRNA expression and accelerated apoptosis of the mesenchyme. These findings suggest that MEST, a gene with a putative mesenchymal cell-derived protein, conceivably plays a role in mammalian metanephric development.
Collapse
Affiliation(s)
- Yashpal S Kanwar
- Department of Pathology, Northwestern University Medical School, Chicago, Illinois 60611, USA.
| | | | | | | | | | | | | |
Collapse
|
16
|
Brunner B, Grützner F, Yaspo ML, Ropers HH, Haaf T, Kalscheuer VM. Molecular cloning and characterization of the Fugu rubripes MEST/COPG2 imprinting cluster and chromosomal localization in Fugu and Tetraodon nigroviridis. Chromosome Res 2001; 8:465-76. [PMID: 11032317 DOI: 10.1023/a:1009263504671] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We isolated Fugu genomic clones using the human MEST (Mesoderm-Specific Transcript) cDNA as probe. Sequence analysis revealed the presence of MEST and three additional genes which show homology to plant DNBP (DNA-Binding Protein), vertebrate COPG2 (Coat Protein Gamma 2), as well as to human and mouse UCN (Urocortin). Structures of Fugu and human MEST, COPG2 and UCN genes are very similar. Since MEST and COPG2 are neighboring genes on human chromosome 7q32, we can conclude that we identified their orthologs and that linkage of these genes is evolutionarily conserved in vertebrates. Unlike human MEST which underlies isoform-specific imprinting and is methylated in a parent-of-origin-specific fashion, the CpG island of the Fugu ortholog is completely methylated. The translation start of Fugu MEST is identical to the non-imprinted human isoform which is in good agreement with the assumption that genomic imprinting is restricted to mammals. Comparative mapping of these genes by fluorescence in-situ hybridization to metaphase chromosomes of Fugu rubripes and Tetraodon nigroviridis showed clear signals on one of the smallest acrocentric chromosomal pairs, which in Fugu, can be easily classified by its unique triangular shape.
Collapse
Affiliation(s)
- B Brunner
- Human Genetics, University Hospital Nijmegen, The Netherlands
| | | | | | | | | | | |
Collapse
|