UB Vice President for Research and Economic Development markup
Xiuqian Mu UB Profile
Ophthalmology
Associate Professor
701 Ellicott Street
Rm B4-307
Buffalo NY 14203
716-881-7463
Export
Publication Types
Co-Authors
  • More Coauthors +
 
Authors: Fuguo Wu, Jonathan E Bard, Julien Kann, Donald Yergeau, Darshan Sapkota, Yichen Ge, Zihua Hu, Jie Wang, Tao Liu, Xiuqian Mu
PubMedId: 33674582
1: Nature communications, vol. 12, 2020
Authors: Sriganesh Ramachandra Rao, Lara A. Skelton, Fuguo Wu, Agnieszka Onysk, Grzegorz Spolnik, Witold Danikiewicz, Mark C. Butler, Delores A. Stacks, Liliana Surmacz, Xiuqian Mu, Ewa Swiezewska, Steven J. Pittler, Steven J. Fliesler
Journal-article: SSRN Electronic Journal, 2020
Authors: Sriganesh Ramachandra Rao, Lara A Skelton, Fuguo Wu, Agnieszka Onysk, Grzegorz Spolnik, Witold Danikiewicz, Mark C Butler, Delores A Stacks, Liliana Surmacz, Xiuqian Mu, Ewa Swiezewska, Steven J Pittler, Steven J Fliesler
PubMedId: 32526701
1: iScience, vol. 23, 2020
Authors: Yichen Ge, Fuguo Wu, Mobin Cheng, Jonathan Bard, Xiuqian Mu
PubMedId: 32741043
1: Developmental dynamics : an official publication of the American Association of Anatomists, 2020
Authors: Mathilde Toch, Audrey Harris, Olivier Schakman, Elena Kondratskaya, Jean-Luc Boulland, Nicolas Dauguet, St?phanie Debrulle, Charlotte Baudouin, Maria Hidalgo-Figueroa, Xiuqian Mu, Alexander Gow, Joel C Glover, Fadel Tissir, Fr?d?ric Clotman
PubMedId: 32764632
1: Scientific reports, vol. 10, 2020
Authors: Mathilde Toch, Audrey Harris, Olivier Schakman, Elena Kondratskaya, Jean-Luc Boulland, Nicolas Dauguet, St?phanie Debrulle, Charlotte Baudouin, Maria Hidalgo-Figueroa, Xiuqian Mu, Alexander Gow, Joel C Glover, Fadel Tissir, Fr?d?ric Clotman
PubMedId: 31969659
1: Scientific reports, vol. 10, 2019
Authors: Qi Zhang, Jamie Zagozewski, Shaohong Cheng, Rajiv Dixit, Shunzhen Zhang, Jimmy de Melo, Xiuqian Mu, William H Klein, Nadean L Brown, Jeffrey T Wigle, Carol Schuurmans, David D Eisenstat
Keywords: Atoh7 Chromatin immunoprecipitation Homeobox In utero electroporation Math5 Mouse
Abstract: Regulated retinal ganglion cell (RGC) differentiation and axonal guidance is required for a functional visual system. Homeodomain and basic helix-loop-helix transcription factors are required for retinogenesis, as well as patterning, differentiation and maintenance of specific retinal cell types. We hypothesized that Dlx1, Dlx2 and Brn3b homeobox genes function in parallel intrinsic pathways to determine RGC fate and therefore generated Dlx1/Dlx2/Brn3b triple-knockout mice. A more severe retinal phenotype was found in the Dlx1/Dlx2/Brn3b-null retinas than was predicted by combining features of the Brn3b single- and Dlx1/Dlx2 double-knockout retinas, including near total RGC loss with a marked increase in amacrine cells in the ganglion cell layer. Furthermore, we discovered that DLX1 and DLX2 function as direct transcriptional activators of Brn3b expression. Knockdown of Dlx2 expression in primary embryonic retinal cultures and Dlx2 gain of function in utero strongly support that DLX2 is both necessary and sufficient for Brn3b expression in vivo We suggest that ATOH7 specifies RGC-committed progenitors and that Dlx1 and Dlx2 function both downstream of ATOH7 and in parallel, but cooperative, pathways that involve regulation of Brn3b expression to determine RGC fate.
PubMedId: 28356311
Journal-article: Development (Cambridge, England), vol. 144, 2016
Authors: Fuguo Wu, Tadeusz J Kaczynski, Santhosh Sethuramanujam, Renzhong Li, Varsha Jain, Malcolm Slaughter, Xiuqian Mu
Keywords: cell fate specification gene regulation neural development retinal development transcription factors
Abstract: As with other retinal cell types, retinal ganglion cells (RGCs) arise from multipotent retinal progenitor cells (RPCs), and their formation is regulated by a hierarchical gene-regulatory network (GRN). Within this GRN, three transcription factors--atonal homolog 7 (Atoh7), POU domain, class 4, transcription factor 2 (Pou4f2), and insulin gene enhancer protein 1 (Isl1)--occupy key node positions at two different stages of RGC development. Atoh7 is upstream and is required for RPCs to gain competence for an RGC fate, whereas Pou4f2 and Isl1 are downstream and regulate RGC differentiation. However, the genetic and molecular basis for the specification of the RGC fate, a key step in RGC development, remains unclear. Here we report that ectopic expression of Pou4f2 and Isl1 in the Atoh7-null retina using a binary knockin-transgenic system is sufficient for the specification of the RGC fate. The RGCs thus formed are largely normal in gene expression, survive to postnatal stages, and are physiologically functional. Our results indicate that Pou4f2 and Isl1 compose a minimally sufficient regulatory core for the RGC fate. We further conclude that during development a core group of limited transcription factors, including Pou4f2 and Isl1, function downstream of Atoh7 to determine the RGC fate and initiate RGC differentiation.
PubMedId: 25775587
Journal-article: Proceedings of the National Academy of Sciences of the United States of America, vol. 112, 2015
Authors: Xiuqian Mu, Darshan Sapkota
PubMedId: 26199604
Journal-article: Neural Regeneration Research, vol. 10, pp. 899, 2015
Authors: Zhiguang Gao, Chai-An Mao, Ping Pan, Xiuqian Mu, William H Klein
Keywords: Atoh7/Math5 eyes absent homolog2 gene regulatory network retinal ganglion cells retinal progenitor cells
Abstract: The bHLH transcription factor ATOH7 (Math5) is essential for establishing retinal ganglion cell (RGC) fate. However, Atoh7-expressing retinal progenitor cells (RPCs) can give rise to all retinal cell types, suggesting that other factors are involved in specifying RGCs. The basis by which a subpopulation of Atoh7-expressing RPCs commits to an RGC fate remains uncertain but is of critical importance to retinal development since RGCs are the earliest cell type to differentiate. To better understand the regulatory mechanisms leading to cell-fate specification, a binary genetic system was generated to specifically label Atoh7-expressing cells with green fluorescent protein (GFP). Fluorescence-activated cell sorting (FACS)-purified GFP(+) and GFP(-) cells were profiled by RNA-seq. Here, we identify 1497 transcripts that were differentially expressed between the two RPC populations. Pathway analysis revealed diminished growth factor signaling in Atoh7-expressing RPCs, indicating that these cells had exited the cell cycle. In contrast, axon guidance signals were enriched, suggesting that axons of Atoh7-expressing RPCs were already making synaptic connections. Notably, many genes enriched in Atoh7-expressing RPCs encoded transcriptional regulators, and several were direct targets of ATOH7, including, and unexpectedly, Ebf3 and Eya2. We present evidence for a Pax6-Atoh7-Eya2 pathway that acts downstream of Atoh7 but upstream of differentiation factor Pou4f2. EYA2 is a protein phosphatase involved in protein-protein interactions and posttranslational regulation. These properties, along with Eya2 as an early target gene of ATOH7, suggest that EYA2 functions in RGC specification. Our results expand current knowledge of the regulatory networks operating in Atoh7-expressing RPCs and offer new directions for exploring the earliest aspects of retinogenesis.
PubMedId: 24799426
Journal-article: Developmental neurobiology, vol. 74, 2014
Authors: Darshan Sapkota, Hemabindu Chintala, Fuguo Wu, Steven J Fliesler, Zihua Hu, Xiuqian Mu
Keywords: cell fate determination gene regulation neural development retinal development transcription factors
Abstract: Previously, we have shown that Onecut1 (Oc1) and Onecut2 (Oc2) are expressed in retinal progenitor cells, developing retinal ganglion cells (RGCs), and horizontal cells (HCs). However, in Oc1-null mice, we only observed an 80% reduction in HCs, but no defects in other cell types. We postulated that the lack of defects in other cell types in Oc1-null retinas was a result of redundancy with Oc2. To test this theory, we have generated Oc2-null mice and now show that their retinas also only have defects in HCs, with a 50% reduction in their numbers. However, when both Oc1 and Oc2 are knocked out, the retinas exhibit more profound defects in the development of all early retinal cell types, including completely failed genesis of HCs, compromised generation of cones, reduced production (by 30%) of RGCs, and absence of starburst amacrine cells. Cone subtype diversification and RGC subtype composition also were affected in the double-null retina. Using RNA-Seq expression profiling, we have identified downstream genes of Oc1 and Oc2, which not only confirms the redundancy between the two factors and renders a molecular explanation for the defects in the double-null retinas, but also shows that the onecut factors suppress the production of the late cell type, rods, indicating that the two factors contribute to the competence of retinal progenitor cells for the early retinal cell fates. Our results provide insight into how onecut factors regulate the creation of cellular diversity in the retina and, by extension, in the central nervous system in general.
PubMedId: 25228773
Journal-article: Proceedings of the National Academy of Sciences of the United States of America, vol. 111, 2014
Authors: Fuguo Wu, Renzhong Li, Yumiko Umino, Tadeusz J Kaczynski, Darshan Sapkota, Shengguo Li, Mengqing Xiang, Steven J Fliesler, David M Sherry, Maureen Gannon, Eduardo Solessio, Xiuqian Mu
Abstract: Horizontal cells are interneurons that synapse with photoreceptors in the outer retina. Their genesis during development is subject to regulation by transcription factors in a hierarchical manner. Previously, we showed that Onecut 1 (Oc1), an atypical homeodomain transcription factor, is expressed in developing horizontal cells (HCs) and retinal ganglion cells (RGCs) in the mouse retina. Herein, by knocking out Oc1 specifically in the developing retina, we show that the majority (80%) of HCs fail to form during early retinal development, implying that Oc1 is essential for HC genesis. However, no other retinal cell types, including RGCs, were affected in the Oc1 knock-out. Analysis of the genetic relationship between Oc1 and other transcription factor genes required for HC development revealed that Oc1 functions downstream of FoxN4, in parallel with Ptf1a, but upstream of Lim1 and Prox1. By in utero electroporation, we found that Oc1 and Ptf1a together are not only essential, but also sufficient for determination of HC fate. In addition, the synaptic connections in the outer plexiform layer are defective in Oc1-null mice, and photoreceptors undergo age-dependent degeneration, indicating that HCs are not only an integral part of the retinal circuitry, but also are essential for the survival of photoreceptors. In sum, these results demonstrate that Oc1 is a critical determinant of HC fate, and reveal that HCs are essential for photoreceptor viability, retinal integrity, and normal visual function.
PubMedId: 23926259
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 33, 2013
Authors: Renzhong Li, Fuguo Wu, Raili Ruonala, Darshan Sapkota, Zihua Hu, Xiuqian Mu
Abstract: Precise regulation of gene expression during biological processes, including development, is often achieved by combinatorial action of multiple transcription factors. The mechanisms by which these factors collaborate are largely not known. We have shown previously that Isl1, a Lim-Homeodomain transcription factor, and Pou4f2, a class IV POU domain transcription factor, co-regulate a set of genes required for retinal ganglion cell (RGC) differentiation. Here we further explore how these two factors interact to precisely regulate gene expression during RGC development. By GST pulldown assays, co-immunoprecipitation, and electrophoretic mobility shift assays, we show that Isl1 and Pou4f2 form a complex in vitro and in vivo, and identify the domains within these two proteins that are responsible for this interaction. By luciferase assay, in situ hybridization, and RNA-seq, we further demonstrate that the two factors contribute quantitatively to gene expression in the developing RGCs. Although each factor alone can activate gene expression, both factors are required to achieve optimal expression levels. Finally, we discover that Isl1 and Pou4f2 can interact with other POU and Lim-Homeodomain factors respectively, indicating the interactions between these two classes of transcription factors are prevalent in development and other biological processes.
PubMedId: 24643061
Journal-article: PloS one, vol. 9, 2013
Authors: Melody Shi, Sumit R Kumar, Oluwaseyi Motajo, Friedrich Kretschmer, Xiuqian Mu, Tudor C Badea
Abstract: Visual information is conveyed from the retina to the brain via 15-20 Retinal Ganglion Cell (RGC) types. The developmental mechanisms by which RGC types acquire their distinct molecular, morphological, physiological and circuit properties are essentially unknown, but may involve combinatorial transcriptional regulation. Brn3 transcription factors are expressed in RGCs from early developmental stages, and are restricted in adults to distinct, partially overlapping populations of RGC types. Previously, we described cell autonomous effects of Brn3b (Pou4f2) and Brn3a (Pou4f1) on RGC axon and dendrites development. We now have investigated genetic interactions between Brn3 transcription factors with respect to RGC development, by crossing conventional knock-out alleles of each Brn3 gene with conditional knock-in reporter alleles of a second Brn3 gene, and analyzing the effects of single or double Brn3 knockouts on RGC survival and morphology. We find that Brn3b loss results in axon defects and dendritic arbor area and lamination defects in Brn3a positive RGCs, and selectively affects survival and morphology of specific Brn3c (Pou4f3) positive RGC types. Brn3a and Brn3b interact synergistically to control RGC numbers. Melanopsin positive ipRGCs are resistant to combined Brn3 loss but are under the transcriptional control of Isl1, expanding the combinatorial code of RGC specification. Taken together these results complete our knowledge on the mechanisms of transcriptional control of RGC type specification. They demonstrate that Brn3b is required for the correct development of more RGC cell types than suggested by its expression pattern in the adult, but that several cell types, including some Brn3a, Brn3c or Melanopsin positive RGCs are Brn3b independent.
PubMedId: 24116103
Journal-article: PloS one, vol. 8, 2013
Authors: Lisa A Ehrman, Xiuqian Mu, Ronald R Waclaw, Yutaka Yoshida, Charles V Vorhees, William H Klein, Kenneth Campbell
Keywords: caudate-putamen direct pathway indirect pathway
Abstract: The mammalian striatum controls the output of the basal ganglia via two distinct efferent pathways, the direct (i.e., striatonigral) and the indirect (i.e., striatopallidal) pathways. The LIM homeodomain transcription factor Islet1 (Isl1) is expressed in a subpopulation of striatal progenitors; however, its specific role in striatal development remains unknown. Our genetic fate-mapping results show that Isl1-expressing progenitors give rise to striatal neurons belonging to the striatonigral pathway. Conditional inactivation of Isl1 in the telencephalon resulted in a smaller striatum with fewer striatonigral neurons and reduced projections to the substantia nigra. Additionally, conditional inactivation in the ventral forebrain (including both the telencephalon and diencephalon) revealed a unique role for Isl1 in diencephalic cells bordering the internal capsule for the normal development of the striatonigral pathway involving PlexinD1-Semaphorin 3e (Sema3e) signaling. Finally, Isl1 conditional mutants displayed a hyperlocomotion phenotype, and their locomotor response to psychostimulants was significantly blunted, indicating that the alterations in basal ganglia circuitry contribute to these mutant behaviors.
PubMedId: 24082127
Journal-article: Proceedings of the National Academy of Sciences of the United States of America, vol. 110, 2013
Authors: Chai-An Mao, Jang-Hyeon Cho, Jing Wang, Zhiguang Gao, Ping Pan, Wen-Wei Tsai, Xiuqian Mu, Laura J. Frishman, William H. Klein
Journal-article: Development, vol. 140, pp. 2849-2849, 2013
Authors: Fuguo Wu, Darshan Sapkota, Renzhong Li, Xiuqian Mu
Abstract: Our current study focuses on the expression of two members of the onecut transcription factor family, Onecut1 (Oc1) and Onecut2 (Oc2), in the developing mouse retina. By immunofluorescence staining, we found that Oc1 and Oc2 had very similar expression patterns throughout retinal development. Both factors started to be expressed in the retina at around embryonic day (E) 11.5. At early stages (E11.5 and E12.5), they were expressed in both the neuroblast layer (NBL) and ganglion cell layer (GCL). As development progressed (from E14.5 to postnatal day [P] 0), expression diminished in the retinal progenitor cells and became more restricted to the GCL. By P5, Oc1 and Oc2 were expressed at very low levels in the GCL. By co-labeling with transcription factors known to be involved in retinal ganglion cell (RGC) development, we found that Oc1 and Oc2 had extensive overlap with Math5 in the NBL, and that they completely overlapped with Pou4f2 and Isl1 in the GCL, but only partially in the NBL. Co-labeling of Oc1 with cell cycle markers confirmed that Oc1 was expressed in both proliferating retinal progenitors and postmitotic retinal cells. In addition, we demonstrated that expression of Oc1 and Oc2 did not require Math5, Isl1, or Pou4f2. Thus, Oc1 and Oc2 may regulate the formation of RGCs in a pathway independent of Math5, Pou4f2, and Isl1. Furthermore, we showed that Oc1 and Oc2 were expressed in both developing and mature horizontal cells (HCs). Therefore the two factors may also function in the genesis and maintenance of HCs.
PubMedId: 21830221
Journal-article: The Journal of comparative neurology, vol. 520, 2011
Authors: Malia M Edwards, D Scott McLeod, Renzhong Li, Rhonda Grebe, Imran Bhutto, Xiuqian Mu, Gerard A Lutty
Abstract: Retinal vascular development is a complex process that is not yet fully understood. The majority of research in this area has focused on astrocytes and the template they form in the inner retina, which precedes endothelial cells in the mouse retina. In humans and dogs, however, astrocyte migration follows behind development of blood vessels, suggesting that other cell types may guide this process. One such cell type is the ganglion cell, which differentiates before blood vessel formation and lies adjacent to the primary retinal vascular plexus. The present study investigated the potential role played by ganglion cells in vascular development using Math5(-/-) mice. It has previously been reported that Math5 regulates the differentiation of ganglion cells and Math5(-/-) mice have a 95% reduction in these cells. The development of blood vessels and glia was investigated using Griffonia simplicifolia isolectin B4 labeling and GFAP immunohistochemistry, respectively. JB-4 analysis demonstrated that the hyaloid vessels arose from choriovitreal vessels adjacent to the optic nerve area. As previously reported, Math5(-/-) mice had a rudimentary optic nerve. The primary retinal vessels did not develop post-natally in the Math5(-/-) mice, however, branches of the hyaloid vasculature eventually dove into the retina and formed the inner retinal capillary networks. An astrocyte template only formed in some areas of the Math5(-/-) retina. In addition, GFAP(+) Mller cells were seen throughout the retina that had long processes wrapped around the hyaloid vessels. Transmission electron microscopy confirmed Mller cell abnormalities and revealed disruptions in the inner limiting membrane. The present data demonstrates that the loss of ganglion cells in the Math5(-/-) mice is associated with a lack of retinal vascular development.
PubMedId: 22200487
Journal-article: Experimental eye research, vol. 96, 2011
Authors: Darshan Sapkota, Fuguo Wu, Xiuqian Mu
PubMedId: 22019371
Journal-article: Experimental eye research, vol. 93, 2011
Authors: Takae Kiyama, Chai-An Mao, Jang-Hyeon Cho, Xueyao Fu, Ping Pan, Xiuqian Mu, William H Klein
Abstract: Retinal progenitor cells (RPCs) are programmed early in development to acquire the competence for specifying the seven retinal cell types. Acquiring competence is a complex spatiotemporal process that is still only vaguely understood. Here, our objective was to more fully understand the mechanisms by which RPCs become competent for specifying a retinal ganglion cell (RGC) fate. RGCs are the first retinal cell type to differentiate and their abnormal development leads to apoptosis and optic nerve degeneration. Previous work demonstrated that the paired domain factor Pax6 and the bHLH factor Atoh7 are required for RPCs to specify RGCs. RGC commitment is marked by the expression of the Pou domain factor Pou4f2 and the Lim domain factor Isl1. We show that three RPC subpopulations can specify RGCs: Atoh7-expressing RPCs, Neurod1-expressing RPCs, and Atoh7-Neurod1-expressing RPCs. All three RPC subpopulations were highly interspersed throughout retinal development, although each subpopulation maintained a distinct temporal pattern. Most, but not all, RPCs from each subpopulation were postmitotic. Atoh7-Neurod1 double knockout mice were generated and double-mutant retinas revealed an unexpected role for Neurod1 in specifying RGC fate. We conclude that RPCs have a complex regulatory gene expression program in which they acquire competence using highly integrated mechanisms.
PubMedId: 20951721
Journal-article: Vision research, vol. 51, 2010
Authors: Xueyao Fu, Takae Kiyama, Renzhong Li, Mark Russell, William H Klein, Xiuqian Mu
Abstract: Although immunological detection of proteins is used extensively in retinal development, studies are often impeded because antibodies against crucial proteins cannot be generated or are not readily available. Here, we overcome these limitations by constructing genetically engineered alleles for Math5 and Pou4f2, two genes required for retinal ganglion cell (RGC) development. Sequences encoding a peptide epitope from haemagglutinin (HA) were added to Math5 or Pou4f2 in frame to generate Math5(HA) and Pou4f2(HA) alleles. We demonstrate that the tagged alleles recapitulated the wild-type expression patterns of the two genes, and that the tags did not interfere with the function of the cognate proteins. In addition, by co-staining, we found that Math5 and Pou4f2 were transiently co-expressed in newly born RGCs, unequivocally demonstrating that Pou4f2 is immediately downstream of Math5 in RGC formation. The epitope-tagged alleles provide new and useful tools for analyzing gene regulatory networks underlying RGC development.
PubMedId: 19459208
Journal-article: Developmental dynamics : an official publication of the American Association of Anatomists, vol. 238, 2009
Authors: Jang-Hyeon Cho, Xiuqian Mu, Steven W Wang, William H Klein
Abstract: Despite the magnitude of the problem, no effective treatments exist to prevent retinal ganglion cell (RGC) death and optic nerve degeneration from occurring in diseases affecting the human eye. Animal models currently available for developing treatment strategies suffer from cumbersome procedures required to induce RGC death or rely on mutations that induce defects in developing retinas rather than in mature retinas of adults. Our objective was to develop a robust genetically engineered adult mouse model for RGC loss and optic nerve degeneration based on genetic ablation. To achieve this, we took advantage of Pou4f2 (Brn3b), a gene activated immediately as RGCs begin to differentiate and expressed throughout life. We generated adult mice whose genomes harbored a conditional Pou4f2 allele containing a floxed-lacZ-stop-diphtheria toxin A cassette and a CAGG-Cre-ER transgene. In this bigenic model, Cre recombinase is fused to a modified estrogen nuclear receptor in which the estrogen-binding domain binds preferentially to the estrogen agonist tamoxifen rather than to endogenous estradiol. Upon binding to the estrogen-binding domain, tamoxifen derepresses Cre recombinase, leading to the efficient genomic deletion of the floxed-lacZ-stop DNA sequence and expression of diphtheria toxin A. Tamoxifen administered to adult mice at different ages by intraperitoneal injection led to rapid RGC loss, reactive gliosis, progressive degradation of the optic nerve over a period of several months, and visual impairment. Perhaps more reflective of human disease, partial loss of RGCs was achieved by modulating the tamoxifen treatment. Especially relevant for RGC death and optic nerve degeneration in human retinal pathologies, RGC-ablated retinas maintained their structural integrity, and other retinal neurons and their connections in the inner and outer plexiform layers appeared unaffected by RGC ablation. These events are hallmarks of progressive optic nerve degeneration observed in human retinal pathologies and demonstrate the validity of this model for use in developing stem cell therapies for replacing dead RGCs with healthy ones.
PubMedId: 19109949
Journal-article: Experimental eye research, vol. 88, 2008
Authors: Xiuqian Mu, Xueyao Fu, Phillip D Beremand, Terry L Thomas, William H Klein
Abstract: Understanding gene regulatory networks (GRNs) that control neuronal differentiation will provide systems-level perspectives on neurogenesis. We have previously constructed a model for a GRN in retinal ganglion cell (RGC) differentiation in which four hierarchical tiers of transcription factors ultimately control the expression of downstream terminal genes. Math5 occupies a central node in the hierarchy because it is essential for the formation of RGCs and the expression of the immediate downstream factor Pou4f2. Based on its expression, we also proposed that Isl1, a LIM-homeodomain factor, functions in parallel with Pou4f2 and downstream of Math5 in the RGC GRN. To determine whether this was the case, a conditional Isl1 allele was generated and deleted specifically in the developing retina. Although RGCs formed in Isl1-deleted retinas, most underwent apoptosis, and few remained at later stages. By microarray analysis, we identified a distinct set of genes whose expression depended on Isl1. These genes are all downstream of Math5, and some of them, but not all, also depend on Pou4f2. Additionally, Isl1 was required for the sustained expression of Pou4f2, suggesting that Isl1 positively regulates Pou4f2 after Math5 levels are diminished. The results demonstrate an essential role for Isl1 in RGC development and reveal two distinct but intersecting branches of the RGC GRN downstream of Math5, one directed by Pou4f2 and the other by Isl1. They also reveal that identical RGC expression patterns are achieved by different combinations of divergent inputs from upstream transcription factors.
PubMedId: 18460603
Journal-article: Proceedings of the National Academy of Sciences of the United States of America, vol. 105, 2008
Authors: Przemyslaw Sapieha, Mirna Sirinyan, David Hamel, Karine Zaniolo, Jean-S??bastien Joyal, Jang-Hyeon Cho, Jean-Claude Honor??, Elsa Kermorvant-Duchemin, Daya R Varma, Sophie Tremblay, Martin Leduc, Lenka Rihakova, Pierre Hardy, William H Klein, Xiuqian Mu, Orval Mamer, Pierre Lachapelle, Adriana Di Polo, Christian Beaus??jour, Gregor Andelfinger, Grant Mitchell, Florian Sennlaub, Sylvain Chemtob
PubMedId: 18836459
Journal-article: Nature Medicine, vol. 14, pp. 1067-1076, 2008
Authors: R.D. Burke, L.M. Angerer, M.R. Elphick, G.W. Humphrey, S. Yaguchi, T. Kiyama, S. Liang, X. Mu, C. Agca, W.H. Klein, B.P. Brandhorst, M. Rowe, K. Wilson, A.M. Churcher, J.S. Taylor, N. Chen, G. Murray, D. Wang, D. Mellott, R. Olinski, F. Hallb????k, M.C. Thorndyke
PubMedId: 16965768
Journal-article: Developmental Biology, vol. 300, pp. 434-460, 2006
Authors: , E. Sodergren, G. M. Weinstock, E. H Davidson, R. A. Cameron, R. A. Gibbs, R. C. Angerer, L. M. Angerer, M. I. Arnone, D. R. Burgess, R. D. Burke, J. A. Coffman, M. Dean, M. R. Elphick, C. A. Ettensohn, K. R. Foltz, A. Hamdoun, R. O. Hynes, W. H. Klein, W. Marzluff, D. R. McClay, R. L. Morris, A. Mushegian, J. P. Rast, L. C. Smith, M. C. Thorndyke, V. D. Vacquier, G. M. Wessel, G. Wray, L. Zhang, C. G. Elsik, O. Ermolaeva, W. Hlavina, G. Hofmann, P. Kitts, M. J. Landrum, A. J. Mackey, D. Maglott, G. Panopoulou, A. J. Poustka, K. Pruitt, V. Sapojnikov, X. Song, A. Souvorov, V. Solovyev, Z. Wei, C. A. Whittaker, K. Worley, K. J. Durbin, Y. Shen, O. Fedrigo, D. Garfield, R. Haygood, A. Primus, R. Satija, T. Severson, M. L. Gonzalez-Garay, A. R. Jackson, A. Milosavljevic, M. Tong, C. E. Killian, B. T. Livingston, F. H. Wilt, N. Adams, R. Belle, S. Carbonneau, R. Cheung, P. Cormier, B. Cosson, J. Croce, A. Fernandez-Guerra, A.-M. Geneviere, M. Goel, H. Kelkar, J. Morales, O. Mulner-Lorillon, A. J. Robertson, J. V. Goldstone, B. Cole, D. Epel, B. Gold, M. E. Hahn, M. Howard-Ashby, M. Scally, J. J. Stegeman, E. L. Allgood, J. Cool, K. M. Judkins, S. S. McCafferty, A. M. Musante, R. A. Obar, A. P. Rawson, B. J. Rossetti, I. R. Gibbons, M. P. Hoffman, A. Leone, S. Istrail, S. C. Materna, M. P. Samanta, V. Stolc, W. Tongprasit, Q. Tu, K.-F. Bergeron, B. P. Brandhorst, J. Whittle, K. Berney, D. J. Bottjer, C. Calestani, K. Peterson, E. Chow, Q. A. Yuan, E. Elhaik, D. Graur, J. T. Reese, I. Bosdet, S. Heesun, M. A. Marra, J. Schein, M. K. Anderson, V. Brockton, K. M. Buckley, A. H. Cohen, S. D. Fugmann, T. Hibino, M. Loza-Coll, A. J. Majeske, C. Messier, S. V. Nair, Z. Pancer, D. P. Terwilliger, C. Agca, E. Arboleda, N. Chen, A. M. Churcher, F. Hallbook, G. W. Humphrey, M. M. Idris, T. Kiyama, S. Liang, D. Mellott, X. Mu, G. Murray, R. P. Olinski, F. Raible, M. Rowe, J. S. Taylor, K. Tessmar-Raible, D. Wang, K. H. Wilson, S. Yaguchi, T. Gaasterland, B. E. Galindo, H. J. Gunaratne, C. Juliano, M. Kinukawa, G. W. Moy, A. T. Neill, M. Nomura, M. Raisch, A. Reade, M. M. Roux, J. L. Song, Y.-H. Su, I. K. Townley, E. Voronina, J. L. Wong, G. Amore, M. Branno, E. R. Brown, V. Cavalieri, V. Duboc, L. Duloquin, C. Flytzanis, C. Gache, F. Lapraz, T. Lepage, A. Locascio, P. Martinez, G. Matassi, V. Matranga, R. Range, F. Rizzo, E. Rottinger, W. Beane, C. Bradham, C. Byrum, T. Glenn, S. Hussain, F. G. Manning, E. Miranda, R. Thomason, K. Walton, A. Wikramanayke, S.-Y. Wu, R. Xu, C. T. Brown, L. Chen, R. F. Gray, P. Y. Lee, J. Nam, P. Oliveri, J. Smith, D. Muzny, S. Bell, J. Chacko, A. Cree, S. Curry, C. Davis, H. Dinh, S. Dugan-Rocha, J. Fowler, R. Gill, C. Hamilton, J. Hernandez, S. Hines, J. Hume, L. Jackson, A. Jolivet, C. Kovar, S. Lee, L. Lewis, G. Miner, M. Morgan, L. V. Nazareth, G. Okwuonu, D. Parker, L.-L. Pu, R. Thorn, R. Wright
Journal-article: Science, vol. 314, pp. 941-952, 2006
Authors: Suzanna E Martin, Xiuqian Mu, William H Klein
Abstract: The POU-domain transcription factor Brn3b/ POU4f2 is an essential regulator of gene expression in mouse retinal ganglion cells. Although Brn3b's importance in the differentiation of these cells has been firmly established, the regions on Brn3b where transcriptional activation and/or repression domains reside are only vaguely defined, and conflicting publications report both activation and repression activities for Brn3b. To clarify its function, we monitored the transcriptional activity of Brn3b and Gal4 DNA-binding domain (DBD)-Brn3b fusion proteins in cotransfection experiments using either Brn3-consensus or Gal4 DNA-binding sites to drive reporter gene expression. At Gal4 DNA-binding sites, transrepression activity mapping to the POU domain within Brn3b's C-terminal region masked any transactivation activity. More detailed experiments revealed that expressing abnormally high levels of POU homeodomain- or other homeodomain-containing sequences caused fortuitous transrepression in the cotransfection assay. To avoid transrepression, Brn3b sequences lacking Brn3b's POU domain were fused to the Gal4 DBD to allow identification of regions that were responsible for transcriptional activation. Considerable transactivation activity was located between amino acid residues 100 and 239, although other regions also had activity. The transactivation domain synergized strongly with another transcription factor, LexA-VP16. At Brn3 DNA-binding sites, full-length Brn3b increased transcription more than 25-fold, and similar activation was observed with the closely related factor Brn3a/POU4f1. No transactivation activity was associated with the C-terminal POU domain-containing portion of Brn3b. The results demonstrate that Brn3b regulates gene expression through the action of a strong transcriptional activation domain within its N-terminal sequence.
PubMedId: 15733064
Journal-article: Differentiation; research in biological diversity, vol. 73, 2005
Authors: Xueyao Fu, Hongxia Sun, William H Klein, Xiuqian Mu
Abstract: During vertebrate retinal development, the seven retinal cell types differentiate sequentially from a single population of retinal progenitor cells (RPCs) and organize themselves into a distinct laminar structure. The purpose of this study was to determine whether beta-catenin, which functions both as a nuclear effector for the canonical Wnt signaling pathway and as a regulator of cell adhesion, is required for retinal neurogenesis or lamination. We used the Cre-loxP system to either eliminate beta-catenin or to express a constitutively active form during retinal neurogenesis. Eliminating beta-catenin did not affect cell differentiation, but did result in the loss of the radial arrangement of RPCs and caused abnormal migration of differentiated neurons. As a result, the laminar structure was massively disrupted in beta-catenin-null retinas, although all retinal cell types still formed. In contrast to other neural tissues, eliminating beta-catenin did not significantly reduce the proliferation rate of RPCs; likewise, activating beta-catenin ectopically in RPCs did not result in overproliferation, but loss of neural retinal identity. These results indicate that beta-catenin is essential during retinal neurogenesis as a regulator of cell adhesion but not as a nuclear effector of the canonical Wnt signaling pathway. The results further imply that retinal lamination and retinal cell differentiation are genetically separable processes.
PubMedId: 16959241
Journal-article: Developmental biology, vol. 299, 2005
Authors: Xiuqian Mu, Xueyao Fu, Hongxia Sun, Phillip D Beremand, Terry L Thomas, William H Klein
Abstract: Math5, a mouse homolog of the Drosophila proneural bHLH transcription factor Atonal, is essential in the developing retina to establish retinal progenitor cell competence for a ganglion cell fate. Elucidating the mechanisms by which Math5 influences progenitor cell competence is crucial for understanding how specification of neuronal cell fate occurs in the retina and it requires knowledge of the downstream target genes that depend on Math5 for their expression. To date, only a handful of genes downstream of Math5 have been identified. To better define the gene network operating downstream of Math5, we used custom-designed microarrays to examine the changes in embryonic retinal gene expression caused by deletion of math5. We identified 270 Math5-dependent genes, including those that were expressed specifically either in progenitor cells or differentiated ganglion cells. The ganglion cell-specific genes included both Brn3b-dependent and Brn3b-independent genes, indicating that Math5 regulates distinct branches of the gene network responsible for retinal ganglion cell differentiation. In math5-null progenitor cells, there was an up-regulation of the proneural genes math3, neuroD, and ngn2, indicating that Math5 suppresses the production of other cell types in addition to promoting retinal ganglion cell formation. The promoter regions of many Math5-dependent genes contained binding sites for REST/NRSF, suggesting that release from general repression in retinal progenitor cells is required for ganglion cell-specific gene activation. The identification of multiple roles for Math5 provides new insights into the gene network that defines progenitor cell competence in the embryonic retina.
PubMedId: 15882586
Journal-article: Developmental biology, vol. 280, 2005
Authors: Shuguang Liang, Sheng Zhao, Xiuqian Mu, Terry Thomas, William H Klein
PubMedId: 15496829
Journal Article: Molecular vision, vol. 10, 2004
Authors: William H. Klein, Xiuqian Mu
Journal-article: Seminars in Cell & Developmental Biology, vol. 15, pp. 51, 2004
Authors: Xiuqian Mu, William H Klein
Abstract: Retinal ganglion cells (RGCs) are the first cell type to be specified during vertebrate retinogenesis. Specification and differentiation of the RGC lineage are a stepwise process involving a hierarchical gene regulatory network. During the past decade, a framework of the network has emerged and key transcriptional regulators have been identified. Pax6, Notch, Ath5, and the Brn3 (Pou4f) factors act at different levels of the regulatory hierarchy. In this review, we summarize the current understanding of the functions of these and other transcriptional factors in the specification and differentiation of the RGC lineage. We emphasize the regulatory relationships among transcription factors at different steps of RGC development. We discuss critical issues that need to be addressed before a complete understanding of the gene regulatory network for RGC development can be achieved. Future directions in RGC development will inevitably rely on combined genetic and genomics approaches.
PubMedId: 15036214
Journal-article: Seminars in cell & developmental biology, vol. 15, 2004
Authors: Xiuqian Mu, Phillip D Beremand, Sheng Zhao, Rashmi Pershad, Hongxia Sun, Ann Scarpa, Shuguang Liang, Terry L Thomas, William H Klein
Abstract: Brn3b/Brn-3.2/POU4f2 is a POU domain transcription factor that is essential for retinal ganglion cell (RGC) differentiation, axonal outgrowth and survival. Our goal was to establish a link between Brn3b and the downstream events leading to RGC differentiation. We sought to determine both the number and types of genes that depend on Brn3b for their expression. RNA probes from wild-type and Brn3b(-/-) E14.5, E16.5 and E18.5 mouse retinas were hybridized to a microarray containing 18,816 retina-expressed cDNAs. At E14.5, we identified 87 genes whose expression was significantly altered in the absence of Brn3b and verified the results by real-time PCR and in situ hybridization. These genes fell into discrete sets that encoded transcription factors, proteins associated with neuron integrity and function, and secreted signaling molecules. We found that Brn3b influenced gene expression in non RGCs of the retina by controlling the expression of secreted signaling molecules such as sonic hedgehog and myostatin/Gdf8. At later developmental stages, additional alterations in gene expression were secondary consequences of aberrant RGC differentiation caused by the absence of Brn3b. Our results demonstrate that a small but crucial fraction of the RGC transcriptome is dependent on Brn3b. The Brn3b-dependent gene sets therefore provide a unique molecular signature for the developing retina.
PubMedId: 14973295
Journal-article: Development (Cambridge, England), vol. 131, 2004
Authors: Xiuqian Mu, Xueyao Fu, Hongxia Sun, Shuguang Liang, Hidetaka Maeda, Laura J Frishman, William H Klein
Abstract: The vertebrate retina develops from an amorphous sheet of dividing retinal progenitor cells (RPCs) through a sequential process that culminates in an exquisitely patterned neural tissue. A current model for retinal development posits that sequential cell-type differentiation is the result of changes in the intrinsic competence state of multipotent RPCs as they advance in time and that the intrinsic changes are influenced by continuous changes in the extracellular environment. Although several studies support the proposition that newly differentiated cells alter the extrinsic state of the developing retina, it is still far from clear what role they play in modifying the extracellular environment and in influencing the properties of RPCs. Here, we specifically ablate retinal ganglion cells (RGCs) as they differentiate, and we determine the impact of RGC absence on retinal development. We find that RGCs are not essential for changing the competence of RPCs, but they are necessary for maintaining sufficient numbers of RPCs by regulating cell proliferation via growth factors. Intrinsic rather than extrinsic factors are likely to play the critical roles in determining retinal cell fate.
PubMedId: 15797020
Journal-article: Current biology : CB, vol. 15, 2004
Authors: Steven W Wang, Xiuqian Mu, William J Bowers, William H Klein
PubMedId: 12507463
Journal Article: Methods (San Diego, Calif.), vol. 28, 2003
Authors: Nicolas O Fortunel, Hasan H Otu, Huck-Hui Ng, Jinhui Chen, Xiuqian Mu, Timothy Chevassut, Xiaoyu Li, Marie Joseph, Charles Bailey, Jacques A Hatzfeld, Antoinette Hatzfeld, Fatih Usta, Vinsensius B Vega, Philip M Long, Towia A Libermann, Bing Lim
PubMedId: 14563990
Journal-article: Science (New York, N.Y.), vol. 302, 2003
Authors: Steven W. Wang, Xiuqian Mu, William J. Bowers, William H. Klein
Journal-article: Methods, vol. 28, pp. 448-456, 2002
Authors: Steven W Wang, Xiuqian Mu, William J Bowers, Dong-Seob Kim, Daniel J Plas, Michael C Crair, Howard J Federoff, Lin Gan, William H Klein
PubMedId: 11807038
Journal Article: Development (Cambridge, England), vol. 129, 2002
Authors: X Mu, S Zhao, R Pershad, T F Hsieh, A Scarpa, S W Wang, R A White, P D Beremand, T L Thomas, L Gan, W H Klein
PubMedId: 11812828
Journal Article: Nucleic acids research, vol. 29, 2002
Authors: X. Mu
Journal-article: Nucleic Acids Research, vol. 29, pp. 4983-4993, 2001
Authors: X Mu, S A Spanos, J Shiloach, A Kimmel
PubMedId: 11493573
Journal Article: Development (Cambridge, England), vol. 128, 2001
Authors: X Mu, B Lee, J M Louis, A R Kimmel
PubMedId: 9716534
Journal Article: Development (Cambridge, England), vol. 125, 1998
Presented by CUBS © 2024