Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Mark G. Carter is active.

Publication


Featured researches published by Mark G. Carter.


Developmental Cell | 2004

Dynamics of Global Gene Expression Changes during Mouse Preimplantation Development

Toshio Hamatani; Mark G. Carter; Alexei A. Sharov; Minoru S.H. Ko

Understanding preimplantation development is important both for basic reproductive biology and for practical applications including regenerative medicine and livestock breeding. Global expression profiles revealed and characterized the distinctive patterns of maternal RNA degradation and zygotic gene activation, including two major transient waves of de novo transcription. The first wave corresponds to zygotic genome activation (ZGA); the second wave, named mid-preimplantation gene activation (MGA), precedes the dynamic morphological and functional changes from the morula to blastocyst stage. Further expression profiling of embryos treated with inhibitors of transcription, translation, and DNA replication revealed that the translation of maternal RNAs is required for the initiation of ZGA. We propose a cascade of gene activation from maternal RNA/protein sets to ZGA gene sets and thence to MGA gene sets. The large number of genes identified as involved in each phase is a first step toward analysis of the complex gene regulatory networks.


Nature Genetics | 2003

Heterozygous disruption of Hic1 predisposes mice to a gender-dependent spectrum of malignant tumors

Wen Yong Chen; Xiaobei Zeng; Mark G. Carter; Craig N. Morrell; Ray Whay Chiu Yen; Manel Esteller; D. Neil Watkins; James G. Herman; Joseph L. Mankowski; Stephen B. Baylin

The gene hypermethylated in cancer-1 (HIC1) encodes a zinc-finger transcription factor that belongs to a group of proteins known as the POZ family. HIC1 is hypermethylated and transcriptionally silent in several types of human cancer. Homozygous disruption of Hic1 impairs development and results in embryonic and perinatal lethality in mice. Here we show that mice disrupted in the germ line for only one allele of Hic1 develop many different spontaneous malignant tumors, including a predominance of epithelial cancers in males and lymphomas and sarcomas in females. The complete loss of Hic1 function in the heterozygous mice seems to involve dense methylation of the promoter of the remaining wild-type allele. We conclude that HIC1 is a candidate tumor-suppressor gene for which loss of function in both mouse and human cancers is associated only with epigenetic modifications.


PLOS Biology | 2003

Transcriptome analysis of mouse stem cells and early embryos.

Alexei A. Sharov; Yulan Piao; Ryo Matoba; Dawood B. Dudekula; Yong Qian; Vincent VanBuren; Geppino Falco; Patrick R. Martin; Carole A. Stagg; Uwem C. Bassey; Yuxia Wang; Mark G. Carter; Toshio Hamatani; Kazuhiro Aiba; Hidenori Akutsu; Lioudmila V. Sharova; Tetsuya S. Tanaka; Wendy L. Kimber; Toshiyuki Yoshikawa; Saied A. Jaradat; Serafino Pantano; Ramaiah Nagaraja; Kenneth R. Boheler; Dennis D. Taub; Richard J. Hodes; Dan L. Longo; David Schlessinger; Jonathan R. Keller; Emily Klotz; Garnett Kelsoe

Understanding and harnessing cellular potency are fundamental in biology and are also critical to the future therapeutic use of stem cells. Transcriptome analysis of these pluripotent cells is a first step towards such goals. Starting with sources that include oocytes, blastocysts, and embryonic and adult stem cells, we obtained 249,200 high-quality EST sequences and clustered them with public sequences to produce an index of approximately 30,000 total mouse genes that includes 977 previously unidentified genes. Analysis of gene expression levels by EST frequency identifies genes that characterize preimplantation embryos, embryonic stem cells, and adult stem cells, thus providing potential markers as well as clues to the functional features of these cells. Principal component analysis identified a set of 88 genes whose average expression levels decrease from oocytes to blastocysts, stem cells, postimplantation embryos, and finally to newborn tissues. This can be a first step towards a possible definition of a molecular scale of cellular potency. The sequences and cDNA clones recovered in this work provide a comprehensive resource for genes functioning in early mouse embryos and stem cells. The nonrestricted community access to the resource can accelerate a wide range of research, particularly in reproductive and regenerative medicine.


PLOS ONE | 2006

Dissecting Oct3/4-Regulated Gene Networks in Embryonic Stem Cells by Expression Profiling

Ryo Matoba; Hitoshi Niwa; Shinji Masui; Satoshi Ohtsuka; Mark G. Carter; Alexei A. Sharov; Minoru S.H. Ko

POU transcription factor Pou5f1 (Oct3/4) is required to maintain ES cells in an undifferentiated state. Here we show that global expression profiling of Oct3/4-manipulated ES cells delineates the downstream target genes of Oct3/4. Combined with data from genome-wide chromatin-immunoprecipitation (ChIP) assays, this analysis identifies not only primary downstream targets of Oct3/4, but also secondary or tertiary targets. Furthermore, the analysis also reveals that downstream target genes are regulated either positively or negatively by Oct3/4. Identification of a group of genes that show both activation and repression depending on Oct3/4 expression levels provides a possible mechanism for the requirement of appropriate Oct3/4 expression to maintain undifferentiated ES cells. As a proof-of-principle study, one of the downstream genes, Tcl1, has been analyzed in detail. We show that Oct3/4 binds to the promoter region of Tcl1 and activates its transcription. We also show that Tcl1 is involved in the regulation of proliferation, but not differentiation, in ES cells. These findings suggest that the global expression profiling of gene-manipulated ES cells can help to delineate the structure and dynamics of gene regulatory networks.


Genome Biology | 2005

Transcript copy number estimation using a mouse whole-genome oligonucleotide microarray

Mark G. Carter; Alexei A. Sharov; Vincent VanBuren; Dawood B. Dudekula; Condie E. Carmack; Charlie Nelson; Minoru S.H. Ko

The ability to quantitatively measure the expression of all genes in a given tissue or cell with a single assay is an exciting promise of gene-expression profiling technology. An in situ-synthesized 60-mer oligonucleotide microarray designed to detect transcripts from all mouse genes was validated, as well as a set of exogenous RNA controls derived from the yeast genome (made freely available without restriction), which allow quantitative estimation of absolute endogenous transcript abundance.


Gene Expression Patterns | 2008

An in situ hybridization-based screen for heterogeneously expressed genes in mouse ES cells

Mark G. Carter; Carole A. Stagg; Geppino Falco; Toshiyuki Yoshikawa; Uwem C. Bassey; Kazuhiro Aiba; Lioudmila V. Sharova; Nabeebi Shaik; Minoru S.H. Ko

We previously reported that Zscan4 showed heterogeneous expression patterns in mouse embryonic stem (ES) cells. To identify genes that show similar expression patterns, we carried out high-throughput in situ hybridization assays on ES cell cultures for 244 genes. Most of the genes are involved in transcriptional regulation, and were selected using microarray-based comparisons of gene expression profiles in ES and embryonal carcinoma (EC) cells versus differentiated cell types. Pou5f1 (Oct4, Oct3/4) and Krt8 (EndoA) were used as controls. Hybridization signals were detected on ES cell colonies for 147 genes (60%). The majority (136 genes) of them showed relatively homogeneous expression in ES cell colonies. However, we found that two genes unequivocally showed Zscan4-like spotted expression pattern (spot-in-colony pattern; Whsc2 and Rhox9). We also found that nine genes showed relatively heterogeneous expression pattern (mosaic-in-colony pattern: Zfp42/Rex1, Rest, Atf4, Pa2g4, E2f2, Nanog, Dppa3/Pgc7/Stella, Esrrb, and Fscn1). Among these genes, Zfp42/Rex1 showed unequivocally heterogeneous expression in individual ES cells prepared by the CytoSpin. These results show the presence of different types or states of cells within ES cell cultures otherwise thought to be undifferentiated and homogeneous, suggesting a previously unappreciated complexity in ES cell cultures.


Stem Cells | 2006

Defining a Developmental Path to Neural Fate by Global Expression Profiling of Mouse Embryonic Stem Cells and Adult Neural Stem/Progenitor Cells

Kazuhiro Aiba; Alexei A. Sharov; Mark G. Carter; Chiara Foroni; Angelo L. Vescovi; Minoru S.H. Ko

To understand global features of gene expression changes during in vitro neural differentiation, we carried out the microarray analysis of embryonic stem cells (ESCs), embryonal carcinoma cells, and adult neural stem/progenitor (NS) cells. Expression profiling of ESCs during differentiation in monolayer culture revealed three distinct phases: undifferentiated ESCs, primitive ectoderm‐like cells, and neural progenitor cells. Principal component (PC) analysis revealed that these cells were aligned on PC1 over the course of 6 days. This PC1 represents approximately 4,000 genes, the expression of which increased with neural commitment/differentiation. Furthermore, NS cells derived from adult brain and their differentiated cells were positioned along this PC axis further away from undifferentiated ESCs than embryonic stem–derived neural progenitors. We suggest that this PC1 defines a path to neural fate, providing a scale for the degree of commitment/differentiation.


PLOS ONE | 2013

MUC1* Ligand, NM23-H1, Is a Novel Growth Factor That Maintains Human Stem Cells in a More Naïve State

Benoit J. Smagghe; Andrew K. Stewart; Mark G. Carter; Laura M. Shelton; Kyle J. Bernier; Eric J. Hartman; Amy K. Calhoun; Vasilios M. Hatziioannou; Gabriele Lillacci; Brian A. Kirk; Brian A. DiNardo; Kenneth S. Kosik; Cynthia Bamdad

We report that a single growth factor, NM23-H1, enables serial passaging of both human ES and iPS cells in the absence of feeder cells, their conditioned media or bFGF in a fully defined xeno-free media on a novel defined, xeno-free surface. Stem cells cultured in this system show a gene expression pattern indicative of a more “naïve” state than stem cells grown in bFGF-based media. NM23-H1 and MUC1* growth factor receptor cooperate to control stem cell self-replication. By manipulating the multimerization state of NM23-H1, we override the stem cells inherent programming that turns off pluripotency and trick the cells into continuously replicating as pluripotent stem cells. Dimeric NM23-H1 binds to and dimerizes the extra cellular domain of the MUC1* transmembrane receptor which stimulates growth and promotes pluripotency. Inhibition of the NM23-H1/MUC1* interaction accelerates differentiation and causes a spike in miR-145 expression which signals a cells exit from pluripotency.


Mammalian Genome | 2004

Klotho gene variation and expression in 20 inbred mouse strains.

Arsun Bektas; Shepherd H. Schurman; Alexei A. Sharov; Mark G. Carter; Harry C. Dietz; Clair A. Francomano

A defect in klotho gene expression in the mouse results in a syndrome that resembles human aging, with greatly shortened lifespan, arteriosclerosis, and defective hearing. In an effort to find functional murine variants of klotho, we sequenced the gene and examined renal expression of the secreted and membrane-bound Klotho isoforms from 16 laboratory-derived and 4 wild-derived inbred strains. Among the laboratory-derived strains, no sequence variation was found in any of the exons or intron–exon boundaries. Among the wild-derived strains, we found 45 sequence variants with six resulting in amino acid substitutions. One wild-derived strain, SPRET/Ei, had four amino acid substitutions and higher levels of the membrane form and total klotho mRNA. In addition, the membrane to secreted klotho expression ratio was elevated in three wildderived strains with amino acid substitutions. Interestingly, SPRET/Ei mice have longer lifespans, decreased atherosclerosis risk factors, and better hearing than almost all other mouse strains.


Stem Cells | 2016

A Primitive Growth Factor, NME7AB, Is Sufficient to Induce Stable Naïve State Human Pluripotency; Reprogramming in This Novel Growth Factor Confers Superior Differentiation

Mark G. Carter; Benoit J. Smagghe; Andrew K. Stewart; J.A. Rapley; E. Lynch; Kyle J. Bernier; K.W. Keating; Vasilios M. Hatziioannou; Eric J. Hartman; Cynthia C. Bamdad

Scientists have generated human stem cells that in some respects mimic mouse naïve cells, but their dependence on the addition of several extrinsic agents, and their propensity to develop abnormal karyotype calls into question their resemblance to a naturally occurring “naïve” state in humans. Here, we report that a recombinant, truncated human NME7, referred to as NME7AB here, induces a stable naïve‐like state in human embryonic stem cells and induced pluripotent stem cells without the use of inhibitors, transgenes, leukemia inhibitory factor (LIF), fibroblast growth factor 2 (FGF2), feeder cells, or their conditioned media. Evidence of a naïve state includes reactivation of the second X chromosome in female source cells, increased expression of naïve markers and decreased expression of primed state markers, ability to be clonally expanded and increased differentiation potential. RNA‐seq analysis shows vast differences between the parent FGF2 grown, primed state cells, and NME7AB converted cells, but similarities to altered gene expression patterns reported by others generating naïve‐like stem cells via the use of biochemical inhibitors. Experiments presented here, in combination with our previous work, suggest a mechanistic model of how human stem cells regulate self‐replication: an early naïve state driven by NME7, which cannot itself limit self‐replication and a later naïve state regulated by NME1, which limits self‐replication when its multimerization state shifts from the active dimer to the inactive hexamer. Stem Cells 2016;34:847–859

Collaboration


Dive into the Mark G. Carter's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexei A. Sharov

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Dawood B. Dudekula

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kazuhiro Aiba

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carole A. Stagg

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ryo Matoba

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Vincent VanBuren

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yong Qian

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Uwem C. Bassey

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge