Network


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

Hotspot


Dive into the research topics where Melanie C. MacNicol is active.

Publication


Featured researches published by Melanie C. MacNicol.


The EMBO Journal | 2002

A novel regulatory element determines the timing of Mos mRNA translation during Xenopus oocyte maturation

Amanda Charlesworth; John A. Ridge; Leslie A. King; Melanie C. MacNicol; Angus M. MacNicol

Progression through vertebrate oocyte maturation requires that pre‐existing, maternally derived mRNAs be translated in a strict temporal order. The mechanism that controls the timing of oocyte mRNA translation is unknown. In this study we show that the early translational induction of the mRNA encoding the Mos proto‐oncogene is mediated through a novel regulatory element within the 3′ untranslated region of the Mos mRNA. This novel element is responsive to the MAP kinase signaling pathway and is distinct from the late acting, cdc2‐responsive, cytoplasmic polyadenylation element. Our findings suggest that the timing of maternal mRNA translation is controlled through signal transduction pathways targeting distinct 3′ UTR mRNA elements.


Aging Cell | 2004

Alterations in the TGFβ signaling pathway in myogenic progenitors with age

Marjorie L. Beggs; Radhakrishnan Nagarajan; Jane M. Taylor-Jones; Greg T. Nolen; Melanie C. MacNicol; Charlotte A. Peterson

Myogenic progenitors in adult muscle are necessary for the repair, maintenance and hypertrophy of post‐mitotic muscle fibers. With age, fat deposition and fibrosis contribute to the decline in the integrity and functional capacity of muscles. In a previous study we reported increased accumulation of lipid in myogenic progenitors obtained from aged mice, accompanied by an up‐regulation of genes involved in adipogenic differentiation. The present study was designed to extend our understanding of how aging affects the fate and gene expression profile of myogenic progenitors. Affymetrix murine U74 Genechip analysis was performed using RNA extracted from myogenic progenitors isolated from adult (8‐month‐old) and aged (24‐month‐old) DBA/2JNIA mice. The cells from the aged animals exhibited major alterations in the expression level of many genes directly or indirectly involved with the TGFβ signaling pathway. Our data indicate that with age, myogenic progenitors acquire the paradoxical phenotype of being both TGFβ activated based on overexpression of TGFβ‐inducible genes, but resistant to the differentiation‐inhibiting effects of exogenous TGFβ. The overexpression of TGFβ‐regulated genes, such as connective tissue growth factor, may play a role in increasing fibrosis in aging muscle.


The EMBO Journal | 2010

Enforcing temporal control of maternal mRNA translation during oocyte cell-cycle progression

Karthik Arumugam; Yiying Wang; Linda L. Hardy; Melanie C. MacNicol; Angus M. MacNicol

Meiotic cell‐cycle progression in progesterone‐stimulated Xenopus oocytes requires that the translation of pre‐existing maternal mRNAs occur in a strict temporal order. Timing of translation is regulated through elements within the mRNA 3′ untranslated region (3′ UTR), which respond to cell cycle‐dependant signalling. One element that has been previously implicated in the temporal control of mRNA translation is the cytoplasmic polyadenylation element (CPE). In this study, we show that the CPE does not direct early mRNA translation. Rather, early translation is directed through specific early factors, including the Musashi‐binding element (MBE) and the MBE‐binding protein, Musashi. Our findings indicate that although the cyclin B5 3′ UTR contains both CPEs and an MBE, the MBE is the critical regulator of early translation. The cyclin B2 3′ UTR contains CPEs, but lacks an MBE and is translationally activated late in maturation. Finally, utilizing antisense oligonucleotides to attenuate endogenous Musashi synthesis, we show that Musashi is critical for the initiation of early class mRNA translation and for the subsequent activation of CPE‐dependant mRNA translation.


Cell Cycle | 2011

Context-dependent regulation of Musashi-mediated mRNA translation and cell cycle regulation.

Melanie C. MacNicol; Chad E. Cragle; Angus M. MacNicol

Musashi-mediated mRNA translational control has been implicated in the promotion of physiological and pathological stem cell proliferation. During self-renewal of mammalian stem cells, Musashi has been proposed to act to repress the translation of mRNAs encoding inhibitors of cell cycle progression. By contrast, in maturing Xenopus oocytes Musashi activates translation of target mRNAs that encode proteins promoting cell cycle progression. The mechanisms directing Musashi to differentially control mRNA translation in mammalian stem cells and Xenopus oocytes is unknown. In this study, we demonstrate that the mechanisms defining Musashi function lie within the cellular context. Specifically, we show that murine Musashi acts as an activator of translation in maturing Xenopus oocytes while Xenopus Musashi functions as a repressor of target mRNA translation in mammalian cells. We further demonstrate that within the context of a primary mammalian neural stem/progenitor cell, Musashi can be converted from a repressor of mRNA translation to an activator of translation in response to extracellular stimuli. We present current models of Musashi-mediated mRNA translational control and discuss possible mechanisms for regulating Musashi function. An understanding of these mechanisms presents exciting possibilities for development of therapeutic targets to control physiological and pathological stem cell proliferation.


Biochemical Society Transactions | 2008

Function and regulation of the mammalian Musashi mRNA translational regulator.

Angus M. MacNicol; Anna Wilczynska; Melanie C. MacNicol

The evolutionarily conserved RNA-binding protein, Musashi, regulates neural stem cell self-renewal. Musashi expression is also indicative of stem cell populations in breast and intestinal tissues and is linked to cell overproliferation in cancers of these tissues. Musashi has been primarily implicated as a repressor of target mRNAs in stem cell populations. However, little is known about the mechanism by which Musashi exerts mRNA translational control or how Musashi function is regulated. Recent findings in oocytes of the frog, Xenopus, indicate an unexpected role for Musashi as an activator of a number of maternal mRNAs during meiotic cell cycle progression. Given the importance of Musashi function in stem cell biology and the implications of aberrant Musashi expression in cancer, it is critical that we understand the molecular processes that regulate Musashi function.


Molecular Reproduction and Development | 2010

Developmental timing of mRNA translation—integration of distinct regulatory elements

Melanie C. MacNicol; Angus M. MacNicol

Targeted mRNA translation is emerging as a critical mechanism to control gene expression during developmental processes. Exciting new findings have revealed a critical role for regulatory elements within the mRNA untranslated regions to direct the timing of mRNA translation. Regulatory elements can be targeted by sequence‐specific binding proteins to direct either repression or activation of mRNA translation in response to developmental signals. As new regulatory elements continue to be identified it has become clear that targeted mRNAs can contain multiple regulatory elements, directing apparently contradictory translational patterns. How is this complex regulatory input integrated? In this review, we focus on a new challenge area—how sequence‐specific RNA binding proteins respond to developmental signals and functionally integrate to regulate the extent and timing of target mRNA translation. We discuss current understanding with a particular emphasis on the control of cell cycle progression that is mediated through a complex interplay of distinct mRNA regulatory elements during Xenopus oocyte maturation. Mol. Reprod. Dev. 77: 662–669, 2010.


Molecular Reproduction and Development | 2012

Autoregulation of Musashi1 mRNA Translation During Xenopus Oocyte Maturation

Karthik Arumugam; Melanie C. MacNicol; Angus M. MacNicol

The mRNA translational control protein, Musashi, plays a critical role in cell fate determination through sequence‐specific interactions with select target mRNAs. In proliferating stem cells, Musashi exerts repression of target mRNAs to promote cell cycle progression. During stem cell differentiation, Musashi target mRNAs are de‐repressed and translated. Recently, we have reported an obligatory requirement for Musashi to direct translational activation of target mRNAs during Xenopus oocyte meiotic cell cycle progression. Despite the importance of Musashi in cell cycle regulation, only a few target mRNAs have been fully characterized. In this study, we report the identification and characterization of a new Musashi target mRNA in Xenopus oocytes. We demonstrate that progesterone‐stimulated translational activation of the Xenopus Musashi1 mRNA is regulated through a functional Musashi binding element (MBE) in the Musashi1 mRNA 3′ untranslated region (3′ UTR). Mutational disruption of the MBE prevented translational activation of Musashi1 mRNA and its interaction with Musashi protein. Further, elimination of Musashi function through microinjection of inhibitory antisense oligonucleotides prevented progesterone‐induced polyadenylation and translation of the endogenous Musashi1 mRNA. Thus, Xenopus Musashi proteins regulate translation of the Musashi1 mRNA during oocyte maturation. Our results indicate that the hierarchy of sequential and dependent mRNA translational control programs involved in directing progression through meiosis are reinforced by an intricate series of nested, positive feedback loops, including Musashi mRNA translational autoregulation. These autoregulatory positive feedback loops serve to amplify a weak initiating signal into a robust commitment for the oocyte to progress through the cell cycle and become competent for fertilization.Mol. Reprod. Dev. 79: 553‐563, 2012.


Journal of Biological Chemistry | 2012

Ringo/Cyclin-dependent Kinase and Mitogen-activated Protein Kinase Signaling Pathways Regulate the Activity of the Cell Fate Determinant Musashi to Promote Cell Cycle Re-entry in Xenopus Oocytes

Karthik Arumugam; Melanie C. MacNicol; Yiying Wang; Chad E. Cragle; Alan J. Tackett; Linda L. Hardy; Angus M. MacNicol

Background: The mechanisms that regulate the activity of the mRNA translational regulator, Musashi, are unknown. Results: Musashi is activated by Ringo/cyclin-dependent kinase and MAP kinase signaling. Conclusion: Musashi-directed mRNA translation induces MAP kinase signaling and establishes a positive feedback loop to amplify Musashi activity. Significance: Musashi activation to promote translation of target mRNAs presents a potential target for the control of pathological cell cycle progression. Cell cycle re-entry during vertebrate oocyte maturation is mediated through translational activation of select target mRNAs, culminating in the activation of mitogen-activated protein kinase and cyclin B/cyclin-dependent kinase (CDK) signaling. The temporal order of targeted mRNA translation is crucial for cell cycle progression and is determined by the timing of activation of distinct mRNA-binding proteins. We have previously shown in oocytes from Xenopus laevis that the mRNA-binding protein Musashi targets translational activation of early class mRNAs including the mRNA encoding the Mos proto-oncogene. However, the molecular mechanism by which Musashi function is activated is unknown. We report here that activation of Musashi1 is mediated by Ringo/CDK signaling, revealing a novel role for early Ringo/CDK function. Interestingly, Musashi1 activation is subsequently sustained through mitogen-activated protein kinase signaling, the downstream effector of Mos mRNA translation, thus establishing a positive feedback loop to amplify Musashi function. The identified regulatory sites are present in mammalian Musashi proteins, and our data suggest that phosphorylation may represent an evolutionarily conserved mechanism to control Musashi-dependent target mRNA translation.


Molecular Reproduction and Development | 2008

Mos 3′ UTR regulatory differences underlie species‐specific temporal patterns of Mos mRNA cytoplasmic polyadenylation and translational recruitment during oocyte maturation

C. Krishna Prasad; Mahendran Mahadevan; Melanie C. MacNicol; Angus M. MacNicol

The Mos proto‐oncogene is a critical regulator of vertebrate oocyte maturation. The maturation‐dependent translation of Mos protein correlates with the cytoplasmic polyadenylation of the maternal Mos mRNA. However, the precise temporal requirements for Mos protein function differ between oocytes of model mammalian species and oocytes of the frog Xenopus laevis. Despite the advances in model organisms, it is not known if the translation of the human Mos mRNA is also regulated by cytoplasmic polyadenylation or what regulatory elements may be involved. We report that the human Mos 3′ untranslated region (3′ UTR) contains a functional cytoplasmic polyadenylation element (CPE) and demonstrate that the endogenous Mos mRNA undergoes maturation‐dependent cytoplasmic polyadenylation in human oocytes. The human Mos 3′ UTR interacts with the human CPE‐binding protein and exerts translational control on a reporter mRNA in the heterologous Xenopus oocyte system. Unlike the Xenopus Mos mRNA, which is translationally activated by an early acting Musashi/polyadenylation response element (PRE)‐directed control mechanism, the translational activation of the human Mos 3′ UTR is dependent on a late acting CPE‐dependent process. Taken together, our findings suggest a fundamental difference in the 3′ UTR regulatory mechanisms controlling the temporal induction of maternal Mos mRNA polyadenylation and translational activation during Xenopus and mammalian oocyte maturation. Mol. Reprod. Dev. 75: 1258–1268, 2008.


Molecular and Cellular Endocrinology | 2002

Early expression of p107 is associated with 3T3-L1 adipocyte differentiation.

Kenian Liu; Yu Guan; Melanie C. MacNicol; Angus M. MacNicol; Robert E. McGehee

In response to hormonal stimulation quiescent 3T3-L1 preadipocyte cells reenter the cell cycle and undergo a mitotic expansion phase prior to terminal differentiation. The cell cycle regulatory proteins p130 and p107 undergo dramatic changes in protein levels within 24 h of differentiation. The role of these proteins in regulating adipocyte mitotic clonal expansion and/or differentiation are unclear. It has recently been demonstrated that adipocyte proliferation can be uncoupled from adipocyte differentiation through the use of the pharmacological MEK inhibitor PD98059 or the tyrosine phosphatase inhibitor, sodium vanadate. We examined the expression of p130 and p107 in stimulated 3T3-L1 cells in the presence of either PD98059, U0126 or sodium vanadate. While inhibition of MEK blocked proliferation, the cells underwent differentiation normally. In contrast, vanadate blocked differentiation without affecting proliferation. Inhibition of MEK did not affect the increase in p107 expression in stimulated cells indicating that induction of p107 is independent of MAP kinase signaling. Vanadate treatment caused a significant delay in p107 expression in the first 24 h following stimulation. Under these conditions, p130 expression was relatively unchanged. Our results indicate that a rapid increase in p107 expression correlates with a commitment to undergo adipocyte differentiation. The data further suggest that the rapid induction of p107 is not required for cellular proliferation during the mitotic clonal expansion phase. Taken together, these findings provide correlative data that implicate p107 in the terminal differentiation, but not proliferation, of quiescent preadipocytes following hormonal stimulation.

Collaboration


Dive into the Melanie C. MacNicol's collaboration.

Top Co-Authors

Avatar

Angus M. MacNicol

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Karthik Arumugam

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Linda L. Hardy

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Angela K. Odle

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Chad E. Cragle

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Gwen V. Childs

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Melody L. Allensworth-James

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Anessa Haney

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Mohsin Md. Syed

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Noor Akhter

University of Arkansas for Medical Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge