Network


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

Hotspot


Dive into the research topics where Linn Gieser is active.

Publication


Featured researches published by Linn Gieser.


PLOS Genetics | 2012

Transcriptional Regulation of Rod Photoreceptor Homeostasis Revealed by In Vivo NRL Targetome Analysis

Hong Hao; Douglas S. Kim; Bernward Klocke; Kory R. Johnson; Kairong Cui; Norimoto Gotoh; Chongzhi Zang; Janina Gregorski; Linn Gieser; Weiqun Peng; Yang C. Fann; Martin Seifert; Keji Zhao; Anand Swaroop

A stringent control of homeostasis is critical for functional maintenance and survival of neurons. In the mammalian retina, the basic motif leucine zipper transcription factor NRL determines rod versus cone photoreceptor cell fate and activates the expression of many rod-specific genes. Here, we report an integrated analysis of NRL-centered gene regulatory network by coupling chromatin immunoprecipitation followed by high-throughput sequencing (ChIP–Seq) data from Illumina and ABI platforms with global expression profiling and in vivo knockdown studies. We identified approximately 300 direct NRL target genes. Of these, 22 NRL targets are associated with human retinal dystrophies, whereas 95 mapped to regions of as yet uncloned retinal disease loci. In silico analysis of NRL ChIP–Seq peak sequences revealed an enrichment of distinct sets of transcription factor binding sites. Specifically, we discovered that genes involved in photoreceptor function include binding sites for both NRL and homeodomain protein CRX. Evaluation of 26 ChIP–Seq regions validated their enhancer functions in reporter assays. In vivo knockdown of 16 NRL target genes resulted in death or abnormal morphology of rod photoreceptors, suggesting their importance in maintaining retinal function. We also identified histone demethylase Kdm5b as a novel secondary node in NRL transcriptional hierarchy. Exon array analysis of flow-sorted photoreceptors in which Kdm5b was knocked down by shRNA indicated its role in regulating rod-expressed genes. Our studies identify candidate genes for retinal dystrophies, define cis-regulatory module(s) for photoreceptor-expressed genes and provide a framework for decoding transcriptional regulatory networks that dictate rod homeostasis.


American Journal of Human Genetics | 2009

Mutations in a BTB-Kelch Protein, KLHL7, Cause Autosomal-Dominant Retinitis Pigmentosa

James S. Friedman; Joseph W. Ray; Naushin Waseem; Kory R. Johnson; Matthew Brooks; Therése Hugosson; Debra K. Breuer; Kari Branham; Daniel S. Krauth; Sara J. Bowne; Lori S. Sullivan; Vesna Ponjavic; Lotta Gränse; Ritu Khanna; Edward H. Trager; Linn Gieser; Dianna Hughbanks-Wheaton; Radu Cojocaru; Noor M. Ghiasvand; Christina Chakarova; Magnus Abrahamson; Harald H H Göring; Andrew R. Webster; David G. Birch; Gonçalo R. Abecasis; Yang C. Fann; Shomi S. Bhattacharya; Stephen P. Daiger; John R. Heckenlively; Sten Andréasson

Retinitis pigmentosa (RP) refers to a genetically heterogeneous group of progressive neurodegenerative diseases that result in dysfunction and/or death of rod and cone photoreceptors in the retina. So far, 18 genes have been identified for autosomal-dominant (ad) RP. Here, we describe an adRP locus (RP42) at chromosome 7p15 through linkage analysis in a six-generation Scandinavian family and identify a disease-causing mutation, c.449G-->A (p.S150N), in exon 6 of the KLHL7 gene. Mutation screening of KLHL7 in 502 retinopathy probands has revealed three different missense mutations in six independent families. KLHL7 is widely expressed, including expression in rod photoreceptors, and encodes a 75 kDa protein of the BTB-Kelch subfamily within the BTB superfamily. BTB-Kelch proteins have been implicated in ubiquitination through Cullin E3 ligases. Notably, all three putative disease-causing KLHL7 mutations are within a conserved BACK domain; homology modeling suggests that mutant amino acid side chains can potentially fill the cleft between two helices, thereby affecting the ubiquitination complexes. Mutations in an identical region of another BTB-Kelch protein, gigaxonin, have previously been associated with giant axonal neuropathy. Our studies suggest an additional role of the ubiquitin-proteasome protein-degradation pathway in maintaining neuronal health and in disease.


Cell Reports | 2016

NRL-Regulated Transcriptome Dynamics of Developing Rod Photoreceptors.

Jung-Woong Kim; Hyun-Jin Yang; Matthew Brooks; Lina Zelinger; Gökhan Karakülah; Norimoto Gotoh; Alexis Boleda; Linn Gieser; Felipe O. Giuste; Dustin Thad Whitaker; Ashley Walton; Rafael Villasmil; Jennifer Barb; Peter J. Munson; Koray Dogan Kaya; Vijender Chaitankar; Tiziana Cogliati; Anand Swaroop

Gene regulatory networks (GRNs) guiding differentiation of cell types and cell assemblies in the nervous system are poorly understood because of inherent complexities and interdependence of signaling pathways. Here, we report transcriptome dynamics of differentiating rod photoreceptors in the mammalian retina. Given that the transcription factor NRL determines rod cell fate, we performed expression profiling of developing NRL-positive (rods) and NRL-negative (S-cone-like) mouse photoreceptors. We identified a large-scale, sharp transition in the transcriptome landscape between postnatal days 6 and 10 concordant with rod morphogenesis. Rod-specific temporal DNA methylation corroborated gene expression patterns. De novo assembly and alternative splicing analyses revealed previously unannotated rod-enriched transcripts and the role of NRL in transcript maturation. Furthermore, we defined the relationship of NRL with other transcriptional regulators and downstream cognate effectors. Our studies provide the framework for comprehensive system-level analysis of the GRN underlying the development of a single sensory neuron, the rod photoreceptor.


Stem cell reports | 2018

Accelerated and Improved Differentiation of Retinal Organoids from Pluripotent Stem Cells in Rotating-Wall Vessel Bioreactors

Tyler DiStefano; Holly Yu Chen; Christopher Panebianco; Koray Dogan Kaya; Matthew Brooks; Linn Gieser; Nicole Y. Morgan; Tom Pohida; Anand Swaroop

Summary Pluripotent stem cells can be differentiated into 3D retinal organoids, with major cell types self-patterning into a polarized, laminated architecture. In static cultures, organoid development may be hindered by limitations in diffusion of oxygen and nutrients. Herein, we report a bioprocess using rotating-wall vessel (RWV) bioreactors to culture retinal organoids derived from mouse pluripotent stem cells. Organoids in RWV demonstrate enhanced proliferation, with well-defined morphology and improved differentiation of neurons including ganglion cells and S-cone photoreceptors. Furthermore, RWV organoids at day 25 (D25) reveal similar maturation and transcriptome profile as those at D32 in static culture, closely recapitulating spatiotemporal development of postnatal day 6 mouse retina in vivo. Interestingly, however, retinal organoids do not differentiate further under any in vitro condition tested here, suggesting additional requirements for functional maturation. Our studies demonstrate that bioreactors can accelerate and improve organoid growth and differentiation for modeling retinal disease and evaluation of therapies.


Science Signaling | 2016

Synergistically acting agonists and antagonists of G protein–coupled receptors prevent photoreceptor cell degeneration

Yu Chen; Grazyna Palczewska; Ikuo Masuho; Songqi Gao; Hui Jin; Zhiqian Dong; Linn Gieser; Matthew Brooks; Philip D. Kiser; Timothy S. Kern; Kirill A. Martemyanov; Anand Swaroop; Krzysztof Palczewski

Systems pharmacology reveals a combination of GPCR-targeted drugs that prevent retinal degeneration. Preventing blindness Loss of photoreceptors in the retina results in visual impairment and eventually blindness. Light can damage the retina through processes that involve G protein–coupled receptors (GPCRs). Chen et al. took a systems pharmacology approach to identify combinations of drugs that activate or inhibit specific GPCRs to prevent light-induced retinal damage in a mouse model of progressive retinal degeneration. This approach identified a photoreceptor-protecting combination of FDA-approved drugs that activated the Gi/o-coupled dopamine receptors D2R and D4R, inhibited the Gs-coupled dopamine receptor D1R, and inhibited the Gq-coupled α1A-adrenergic receptor. This study not only provides a potential therapeutic strategy for preventing blindness due to retinal degeneration but also suggests that systems pharmacology approaches could lead to the discovery of new combinations of available drugs to promote therapeutic changes in signaling networks. Photoreceptor cell degeneration leads to visual impairment and blindness in several types of retinal disease. However, the discovery of safe and effective therapeutic strategies conferring photoreceptor cell protection remains challenging. Targeting distinct cellular pathways with low doses of different drugs that produce a functionally synergistic effect could provide a strategy for preventing or treating retinal dystrophies. We took a systems pharmacology approach to identify potential combination therapies using a mouse model of light-induced retinal degeneration. We showed that a combination of U.S. Food and Drug Administration–approved drugs that act on different G protein (heterotrimeric guanine nucleotide–binding protein)–coupled receptors (GPCRs) exhibited synergistic activity that protected retinas from light-induced degeneration even when each drug was administered at a low dose. In functional assays, the combined effects of these drugs were stimulation of Gi/o signaling by activating the dopamine receptors D2R and D4R, as well as inhibition of Gs and Gq signaling by antagonizing D1R and the α1A-adrenergic receptor ADRA1A, respectively. Moreover, transcriptome analyses demonstrated that such combined GPCR-targeted treatments preserved patterns of retinal gene expression that were more similar to those of the normal retina than did higher-dose monotherapy. Our study thus supports a systems pharmacology approach to identify treatments for retinopathies, an approach that could extend to other complex disorders.


Pharmacological Research | 2017

Transcriptome profiling of NIH3T3 cell lines expressing opsin and the P23H opsin mutant identifies candidate drugs for the treatment of retinitis pigmentosa

Yuanyuan Chen; Matthew Brooks; Linn Gieser; Anand Swaroop; Krzysztof Palczewski

Graphical abstract Figure. No caption available. ABSTRACT Mammalian cells are commonly employed in screening assays to identify active compounds that could potentially affect the progression of different human diseases including retinitis pigmentosa (RP), a class of inherited diseases causing retinal degeneration with compromised vision. Using transcriptome analysis, we compared NIH3T3 cells expressing wildtype (WT) rod opsin with a retinal disease‐causing single P23H mutation. Surprisingly, heterologous expression of WT opsin in NIH3T3 cells caused more than a 2‐fold change in 783 out of 16,888 protein coding transcripts. The perturbed genes encoded extracellular matrix proteins, growth factors, cytoskeleton proteins, glycoproteins and metalloproteases involved in cell adhesion, morphology and migration. A different set of 347 transcripts was either up‐ or down‐regulated when the P23H mutant opsin was expressed suggesting an altered molecular perturbation compared to WT opsin. Transcriptome perturbations elicited by drug candidates aimed at mitigating the effects of the mutant protein revealed that different drugs targeted distinct molecular pathways that resulted in a similar phenotype selected by a cell‐based high‐throughput screen. Thus, transcriptome profiling can provide essential information about the therapeutic potential of a candidate drug to restore normal gene expression in pathological conditions.


Investigative Ophthalmology & Visual Science | 2017

Retinal Degeneration Triggers the Activation of YAP/TEAD in Reactive Müller Cells

Annaïg Hamon; Christel Masson; Juliette Bitard; Linn Gieser; Jerome E. Roger; Muriel Perron

Purpose During retinal degeneration, Müller glia cells respond to photoreceptor loss by undergoing reactive gliosis, with both detrimental and beneficial effects. Increasing our knowledge of the complex molecular response of Müller cells to retinal degeneration is thus essential for the development of new therapeutic strategies. The purpose of this work was to identify new factors involved in Müller cell response to photoreceptor cell death. Methods Whole transcriptome sequencing was performed from wild-type and degenerating rd10 mouse retinas at P30. The changes in mRNA abundance for several differentially expressed genes were assessed by quantitative RT-PCR (RT-qPCR). Protein expression level and retinal cellular localization were determined by western blot and immunohistochemistry, respectively. Results Pathway-level analysis from whole transcriptomic data revealed the Hippo/YAP pathway as one of the main signaling pathways altered in response to photoreceptor degeneration in rd10 retinas. We found that downstream effectors of this pathway, YAP and TEAD1, are specifically expressed in Müller cells and that their expression, at both the mRNA and protein levels, is increased in rd10 reactive Müller glia after the onset of photoreceptor degeneration. The expression of Ctgf and Cyr61, two target genes of the transcriptional YAP/TEAD complex, is also upregulated following photoreceptor loss. Conclusions This work reveals for the first time that YAP and TEAD1, key downstream effectors of the Hippo pathway, are specifically expressed in Müller cells. We also uncovered a deregulation of the expression and activity of Hippo/YAP pathway components in reactive Müller cells under pathologic conditions.


Human Molecular Genetics | 2018

Patient iPSC-derived neural stem cells exhibit phenotypes in concordance with the clinical severity of mucopolysaccharidosis I

Manju Swaroop; Matthew Brooks; Linn Gieser; Anand Swaroop; Wei Zheng

Abstract Mucopolysaccharidosis type I (MPS I) is caused by deficiency of &agr;‐l‐iduronidase (IDUA), a lysosomal enzyme involved in the breakdown and recycling of glycosaminoglycans (GAGs). Although enzyme replacement therapy is available, the efficacy of the treatment for neuropathic manifestations is limited. To facilitate drug discovery and model disease pathophysiology, we generated neural stem cells (NSCs) from MPS I patient‐derived induced pluripotent stem cells (iPSCs). The NSCs exhibited characteristic disease phenotypes with deficiency of IDUA, accumulation of GAGs and enlargement of lysosomes, in agreement with the severity of clinical subgroups of MPS I. Transcriptome profiling of NSCs revealed 429 genes that demonstrated a more extensive change in expression in the most severe Hurler syndrome subgroup compared to the intermediate Hurler‐Scheie or the least severe Scheie syndrome subgroups. Clustering and pathway analysis revealed high concordance of the severity of neurological defects with marked dysregulation of GAG biosynthesis, GAG degradation, lysosomal function and autophagy. Gene ontology (GO) analysis identified a dramatic upregulation of the autophagy pathway, especially in the Hurler syndrome subgroup. We conclude that GAG accumulation in the patient‐derived cells disrupts lysosomal homeostasis, affecting multiple related cellular pathways in response to IDUA deficiency. These dysregulated processes likely lead to enhanced autophagy and progressively severe disease states. Our study provides potentially useful targets for clinical biomarker development, disease diagnosis and prognosis, and drug discovery.


Neurobiology of Aging | 2013

Gene expression changes in aging retinal microglia: relationship to microglial support functions and regulation of activation.

Wenxin Ma; Radu Cojocaru; Norimoto Gotoh; Linn Gieser; Rafael Villasmil; Tiziana Cogliati; Anand Swaroop; Wai T. Wong


Genomics | 1997

A 2.8-Mb clone contig of the multiple endocrine neoplasia type 1 (MEN1) region at 11q13

Siradanahalli C. Guru; Shodimu-Emmanuel Olufemi; Pachiappan Manickam; Christiano Cummings; Linn Gieser; Brian L. Pike; Michael L. Bittner; Yuan Jiang; A. Craig Chinault; Norma J. Nowak; Anna Brzozowska; Judy S. Crabtree; Yingping Wang; Bruce A. Roe; Jane M. Weisemann; Mark S. Boguski; Sunita K. Agarwal; A. Lee Burns; Allen M. Spiegel; Stephen J. Marx; Wendy L. Flejter; Pieter J. de Jong; Francis S. Collins; Settara C. Chandrasekharappa

Collaboration


Dive into the Linn Gieser's collaboration.

Top Co-Authors

Avatar

Anand Swaroop

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Matthew Brooks

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rafael Villasmil

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Tiziana Cogliati

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Radu Cojocaru

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Alexis Boleda

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ashley Walton

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Koray Dogan Kaya

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kory R. Johnson

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge