Network


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

Hotspot


Dive into the research topics where Daniel W. Young is active.

Publication


Featured researches published by Daniel W. Young.


Oncogene | 2004

Runx2 control of organization, assembly and activity of the regulatory machinery for skeletal gene expression

Gary S. Stein; Jane B. Lian; Andre J. Van Wijnen; Janet L. Stein; Martin A. Montecino; Amjad Javed; Sayyed K. Zaidi; Daniel W. Young; Je-Yong Choi; Shirwin M. Pockwinse

We present an overview of Runx involvement in regulatory mechanisms that are requisite for fidelity of bone cell growth and differentiation, as well as for skeletal homeostasis and the structural and functional integrity of skeletal tissue. Runx-mediated control is addressed from the perspective of support for biological parameters of skeletal gene expression. We review recent findings that are consistent with an active role for Runx proteins as scaffolds for integration, organization and combinatorial assembly of nucleic acids and regulatory factors within the three-dimensional context of nuclear architecture.


Reviews in Endocrine & Metabolic Disorders | 2006

Networks and hubs for the transcriptional control of osteoblastogenesis

Jane B. Lian; Gary S. Stein; Amjad Javed; Andre J. Van Wijnen; Janet L. Stein; Martin A. Montecino; Mohammad Q. Hassan; Tripti Gaur; Christopher J. Lengner; Daniel W. Young

We present an overview of the concepts of tissue-specific transcriptional control mechanisms essential for development of the bone cell phenotype. BMP2 induced transcription factors constitute a network of activities and molecular switches for bone development and osteoblast differentiation. Among these regulators are Runx2 (Cbfa1/AML3), the principal osteogenic master gene for bone formation, as well as homeodomain proteins and osterix. Runx2 has multiple regulatory activities, including activation or repression of gene expression, and integration of biological signals from developmental cues, such as BMP/TGFβ, Wnt and Src signaling pathways. Runx2 provides a new paradigm for transcriptional control by functioning as a principal scaffolding protein in nuclear microenvironments to control gene expression in response to physiologic signals (growth factors, cytokines and hormones). The protein serves as a hub for the coordination of activities essential for the expansion and differentiation of osteogenic lineage cells through the formation of co-regulatory protein complexes organized in subnuclear domains. Mechanisms by which Runx2 supports commitment to osteogenesis and determines cell fate involve its retention on mitotic chromosomes. Disruption of a unique protein module, the subnuclear targeting signal of Runx2, has profound effects on osteoblast differentiation and metastasis of cancer cells in the bone microenvironment. Runx2 target genes include regulators of cell growth control, components of the bone extracellular matrix, angiogenesis, and signaling proteins for development of the osteoblast phenotype and bone turnover. The specificity of Runx2 regulatory activities provides a basis for novel therapeutic strategies to correct bone disorders.


Nature | 2007

Mitotic occupancy and lineage-specific transcriptional control of rRNA genes by Runx2

Daniel W. Young; Mohammad Q. Hassan; Jitesh Pratap; Mario Galindo; Sayyed K. Zaidi; Suk Hee Lee; Xiaoqing Yang; Ronglin Xie; Amjad Javed; Jean M. Underwood; Paul S. Furcinitti; Anthony N. Imbalzano; Sheldon Penman; Jeffrey A. Nickerson; Martin A. Montecino; Jane B. Lian; Janet L. Stein; Andre J. Van Wijnen; Gary S. Stein

Regulation of ribosomal RNA genes is a fundamental process that supports the growth of cells and is tightly coupled with cell differentiation. Although rRNA transcriptional control by RNA polymerase I (Pol I) and associated factors is well studied, the lineage-specific mechanisms governing rRNA expression remain elusive. Runt-related transcription factors Runx1, Runx2 and Runx3 establish and maintain cell identity, and convey phenotypic information through successive cell divisions for regulatory events that determine cell cycle progression or exit in progeny cells. Here we establish that mammalian Runx2 not only controls lineage commitment and cell proliferation by regulating genes transcribed by RNA Pol II, but also acts as a repressor of RNA Pol I mediated rRNA synthesis. Within the condensed mitotic chromosomes we find that Runx2 is retained in large discrete foci at nucleolar organizing regions where rRNA genes reside. These Runx2 chromosomal foci are associated with open chromatin, co-localize with the RNA Pol I transcription factor UBF1, and undergo transition into nucleoli at sites of rRNA synthesis during interphase. Ribosomal RNA transcription and protein synthesis are enhanced by Runx2 deficiency that results from gene ablation or RNA interference, whereas induction of Runx2 specifically and directly represses rDNA promoter activity. Runx2 forms complexes containing the RNA Pol I transcription factors UBF1 and SL1, co-occupies the rRNA gene promoter with these factors in vivo, and affects local chromatin histone modifications at rDNA regulatory regions. Thus Runx2 is a critical mechanistic link between cell fate, proliferation and growth control. Our results suggest that lineage-specific control of ribosomal biogenesis may be a fundamental function of transcription factors that govern cell fate.


Journal of Cellular Biochemistry | 2003

Phenotype discovery by gene expression profiling: Mapping of biological processes linked to BMP-2-mediated osteoblast differentiation

Eva Balint; David S. Lapointe; Hicham Drissi; Caroline M. J. van der Meijden; Daniel W. Young; Andre J. Van Wijnen; Janet L. Stein; Gary S. Stein; Jane B. Lian

Understanding physiological control of osteoblast differentiation necessitates characterization of the regulatory signals that initiate the events directing a cell to lineage commitment and establishing competency for bone formation. The bone morphogenetic protein, BMP‐2, a member of the TGFβ superfamily, induces osteoblast differentiation and functions through the Smad signal transduction pathway during in vivo bone formation. However, the molecular targets of BMP‐mediated gene transcription during the process of osteoblast differentiation have not been comprehensively identified. In the present study, BMP‐2 responsive factors involved in the early stages of commitment and differentiation to the osteoblast phenotype were analyzed by microarray gene expression profiling in samples ranging from 1 to 24 h following BMP‐2 dependent differentiation of C2C12 premyoblasts into the osteogenic lineage. A total of 1,800 genes were responsive to BMP‐2 and expression was modulated from 3‐ to 14‐fold for less than 100 genes during the time course. Approximately 50% of these 100 genes are either up‐ or downregulated. Major events associated with phenotypic changes towards the osteogenic lineage were identified from hierarchical and functional clustering analyses. BMP‐2 immediately responsive genes (1–4 h), which exhibited either transient or sustained expression, reflect activation and repression of non‐osseous BMP‐2 developmental systems. This initial response was followed by waves of expression of nuclear proteins and developmental regulatory factors including inhibitors of DNA binding, Runx2, C/EBP, Zn finger binding proteins, forkhead, and numerous homeobox proteins (e.g., CDP/cut, paired, distaless, Hox) which are expressed at characterized stages during osteoblast differentiation. A sequential profile of genes mediating changes in cell morphology, cell growth, and basement membrane formation is observed as a secondary transient early response (2–8 h). Commitment to the osteogenic phenotype is recognized by 8 h, reflected by downregulation of most myogenic‐related genes and induction of a spectrum of signaling proteins and enzymes facilitating synthesis and assembly of an extracellular skeletal environment. These genes included collagens Type I and VI and the small leucine rich repeat family of proteoglycans (e.g., decorin, biglycan, osteomodulin, fibromodulin, and osteoadherin/osteoglycin) that reached peak expression at 24 h. With extracellular matrix development, the bone phenotype was further established from 16 to 24 h by induction of genes for cell adhesion and communication and enzymes that organize the bone ECM. Our microarray analysis resulted in the discovery of a class of genes, initially described in relation to differentiation of astrocytes and oligodendrocytes that are functionally coupled to signals for cellular extensions. They include nexin, neuropilin, latexin, neuroglian, neuron specific gene 1, and Ulip; suggesting novel roles for these genes in the bone microenvironment. This global analysis identified a multistage molecular and cellular cascade that supports BMP‐2‐mediated osteoblast differentiation. J. Cell. Biochem. 89: 401–426, 2003.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Mitotic retention of gene expression patterns by the cell fate-determining transcription factor Runx2

Daniel W. Young; Mohammad Q. Hassan; Xiaoqing Yang; Mario Galindo; Amjad Javed; Sayyed K. Zaidi; Paul S. Furcinitti; David S. Lapointe; Martin A. Montecino; Jane B. Lian; Janet L. Stein; Andre J. Van Wijnen; Gary S. Stein

During cell division, cessation of transcription is coupled with mitotic chromosome condensation. A fundamental biological question is how gene expression patterns are retained during mitosis to ensure the phenotype of progeny cells. We suggest that cell fate-determining transcription factors provide an epigenetic mechanism for the retention of gene expression patterns during cell division. Runx proteins are lineage-specific transcription factors that are essential for hematopoietic, neuronal, gastrointestinal, and osteogenic cell fates. Here we show that Runx2 protein is stable during cell division and remains associated with chromosomes during mitosis through sequence-specific DNA binding. Using siRNA-mediated silencing, mitotic cell synchronization, and expression profiling, we identify Runx2-regulated genes that are modulated postmitotically. Novel target genes involved in cell growth and differentiation were validated by chromatin immunoprecipitation. Importantly, we find that during mitosis, when transcription is shut down, Runx2 selectively occupies target gene promoters, and Runx2 deficiency alters mitotic histone modifications. We conclude that Runx proteins have an active role in retaining phenotype during cell division to support lineage-specific control of gene expression in progeny cells.


Nature Reviews Cancer | 2007

Nuclear microenvironments in biological control and cancer

Sayyed K. Zaidi; Daniel W. Young; Amjad Javed; Jitesh Pratap; Martin A. Montecino; Andre J. Van Wijnen; Jane B. Lian; Janet L. Stein; Gary S. Stein

Nucleic acids and regulatory proteins are compartmentalized in microenvironments within the nucleus. This subnuclear organization may support convergence and the integration of physiological signals for the combinatorial control of gene expression, DNA replication and repair. Nuclear organization is modified in many cancers. There are cancer-related changes in the composition, organization and assembly of regulatory complexes at intranuclear sites. Mechanistic insights into the temporal and spatial organization of machinery for gene expression within the nucleus, which is compromised in tumours, provide a novel platform for diagnosis and therapy.


EMBO Reports | 2005

The dynamic organization of gene-regulatory machinery in nuclear microenvironments

Sayyed K. Zaidi; Daniel W. Young; Je-Yong Choi; Jitesh Pratap; Amjad Javed; Martin A. Montecino; Janet L. Stein; Andre J. Van Wijnen; Jane B. Lian; Gary S. Stein

Nuclear components are functionally linked with the dynamic temporal and spatial compartmentalization, sorting and integration of regulatory information to facilitate its selective use. For example, the subnuclear targeting of transcription factors to punctate sites in the interphase nucleus mechanistically couples chromatin remodelling and the execution of signalling cascades that mediate gene expression with the combinatorial assembly of the regulatory machinery for biological control. In addition, a mitotic cycle of selective partitioning and sequential restoration of the transcriptional machinery provides a basis for the reassembly of regulatory complexes to render progeny cells competent for phenotypic gene expression. When this intranuclear targeting and localization of regulatory proteins is compromised, diseases, such as cancer, can occur. A detailed understanding of this process will provide further options for diagnosis and treatment.


Nature Reviews Genetics | 2010

Mitotic bookmarking of genes: a novel dimension to epigenetic control

Sayyed K. Zaidi; Daniel W. Young; Martin A. Montecino; Jane B. Lian; Andre J. Van Wijnen; Janet L. Stein; Gary S. Stein

Regulatory machinery is focally organized in the interphase nucleus. The information contained in these focal nuclear microenvironments must be inherited during cell division to sustain physiologically responsive gene expression in progeny cells. Recent results suggest that focal mitotic retention of phenotypic transcription factors at promoters together with histone modifications and DNA methylation — a mechanism collectively known as gene bookmarking — is a novel parameter of inherited epigenetic control that sustains cellular identity after mitosis. The epigenetic signatures imposed by bookmarking poise genes for activation or suppression following mitosis. We discuss the implications of phenotypic transcription factor retention on mitotic chromosomes in biological control and disease.


Journal of Biological Chemistry | 2008

Runx2 regulates G protein-coupled signaling pathways to control growth of osteoblast progenitors

Nadiya M. Teplyuk; Mario Galindo; Viktor I. Teplyuk; Jitesh Pratap; Daniel W. Young; David S. Lapointe; Amjad Javed; Janet L. Stein; Jane B. Lian; Gary S. Stein; Andre J. Van Wijnen

Runt-related transcription factor 2 (Runx2) controls lineage commitment, proliferation, and anabolic functions of osteoblasts as the subnuclear effector of multiple signaling axes (e.g. transforming growth factor-β/BMP-SMAD, SRC/YES-YAP, and GROUCHO/TLE). Runx2 levels oscillate during the osteoblast cell cycle with maximal levels in G1. Here we examined what functions and target genes of Runx2 control osteoblast growth. Forced expression of wild type Runx2 suppresses growth of Runx2-/- osteoprogenitors. Point mutants defective for binding to WW domain or SMAD proteins or the nuclear matrix retain this growth regulatory ability. Hence, key signaling pathways are dispensable for growth control by Runx2. However, mutants defective for DNA binding or C-terminal gene repression/activation functions do not block proliferation. Target gene analysis by Affymetrix expression profiling shows that the C terminus of Runx2 regulates genes involved in G protein-coupled receptor signaling (e.g. Rgs2, Rgs4, Rgs5, Rgs16, Gpr23, Gpr30, Gpr54, Gpr64, and Gna13). We further examined the function of two genes linked to cAMP signaling as follows: Gpr30 that is stimulated and Rgs2 that is down-regulated by Runx2. RNA interference of Gpr30 and forced expression of Rgs2 in each case inhibit osteoblast proliferation. Notwithstanding its growth-suppressive potential, our results surprisingly indicate that Runx2 may sensitize cAMP-related G protein-coupled receptor signaling by activating Gpr30 and repressing Rgs2 gene expression in osteoblasts to increase responsiveness to mitogenic signals.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Mitotic partitioning and selective reorganization of tissue-specific transcription factors in progeny cells

Sayyed K. Zaidi; Daniel W. Young; Shirwin M. Pockwinse; Amjad Javed; Jane B. Lian; Janet L. Stein; Andre J. Van Wijnen; Gary S. Stein

Postmitotic gene expression requires restoration of nuclear organization and assembly of regulatory complexes. The hematopoietic and osteogenic Runx (Cbfa/AML) transcription factors are punctately organized in the interphase nucleus and provide a model for understanding the subnuclear organization of tissue-specific regulatory proteins after mitosis. Here we have used quantitative in situ immunofluorescence microscopy and quantitative image analysis to show that Runx factors undergo progressive changes in cellular localization during mitosis while retaining a punctate distribution. In comparison, the acetyl transferase p300 and acetylated histone H4 remain localized with DNA throughout mitosis while the RNA processing factor SC35 is excluded from mitotic chromatin. Subnuclear organization of Runx foci is completely restored in telophase, and Runx proteins are equally partitioned into progeny nuclei. In contrast, subnuclear organization of SC35 is restored subsequent to telophase. Our results show a sequential reorganization of Runx and its coregulatory proteins that precedes restoration of RNA processing speckles. Thus, mitotic partitioning and spatiotemporal reorganization of regulatory proteins together render progeny cells equivalently competent to support phenotypic gene expression.

Collaboration


Dive into the Daniel W. Young's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andre J. Van Wijnen

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amjad Javed

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Jitesh Pratap

Rush University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Mohammad Q. Hassan

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Je-Yong Choi

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Syed A. Ali

University of Massachusetts Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge