Network


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

Hotspot


Dive into the research topics where Wei-Chieh Chiang is active.

Publication


Featured researches published by Wei-Chieh Chiang.


Nature Genetics | 2015

Mutations in the unfolded protein response regulator ATF6 cause the cone dysfunction disorder achromatopsia

Susanne Kohl; Ditta Zobor; Wei-Chieh Chiang; Nicole Weisschuh; Jennifer Staller; Irene Gonzalez Menendez; Stanley Chang; Susanne C. Beck; Marina Garcia Garrido; Vithiyanjali Sothilingam; Mathias W. Seeliger; Franco Stanzial; Francesco Benedicenti; Francesca Inzana; Elise Héon; Ajoy Vincent; Jill Beis; Tim M. Strom; Günther Rudolph; Susanne Roosing; Anneke I. den Hollander; Frans P.M. Cremers; Irma Lopez; Huanan Ren; Anthony T. Moore; Andrew R. Webster; Michel Michaelides; Robert K. Koenekoop; Eberhart Zrenner; Randal J. Kaufman

Achromatopsia (ACHM) is an autosomal recessive disorder characterized by color blindness, photophobia, nystagmus and severely reduced visual acuity. Using homozygosity mapping and whole-exome and candidate gene sequencing, we identified ten families carrying six homozygous and two compound-heterozygous mutations in the ATF6 gene (encoding activating transcription factor 6A), a key regulator of the unfolded protein response (UPR) and cellular endoplasmic reticulum (ER) homeostasis. Patients had evidence of foveal hypoplasia and disruption of the cone photoreceptor layer. The ACHM-associated ATF6 mutations attenuate ATF6 transcriptional activity in response to ER stress. Atf6−/− mice have normal retinal morphology and function at a young age but develop rod and cone dysfunction with increasing age. This new ACHM-related gene suggests a crucial and unexpected role for ATF6A in human foveal development and cone function and adds to the list of genes that, despite ubiquitous expression, when mutated can result in an isolated retinal photoreceptor phenotype.


American Journal of Pathology | 2015

Multiple Mechanisms of Unfolded Protein Response–Induced Cell Death

Nobuhiko Hiramatsu; Wei-Chieh Chiang; Timothy D. Kurt; Christina J. Sigurdson; Jonathan H. Lin

Eukaryotic cells fold and assemble membrane and secreted proteins in the endoplasmic reticulum (ER), before delivery to other cellular compartments or the extracellular environment. Correctly folded proteins are released from the ER, and poorly folded proteins are retained until they achieve stable conformations; irreparably misfolded proteins are targeted for degradation. Diverse pathological insults, such as amino acid mutations, hypoxia, or infection, can overwhelm ER protein quality control, leading to misfolded protein buildup, causing ER stress. To cope with ER stress, eukaryotic cells activate the unfolded protein response (UPR) by increasing levels of ER protein-folding enzymes and chaperones, enhancing the degradation of misfolded proteins, and reducing protein translation. In mammalian cells, three ER transmembrane proteins, inositol-requiring enzyme-1 (IRE1; official name ERN1), PKR-like ER kinase (PERK; official name EIF2AK3), and activating transcription factor-6, control the UPR. The UPR signaling triggers a set of prodeath programs when the cells fail to successfully adapt to ER stress or restore homeostasis. ER stress and UPR signaling are implicated in the pathogenesis of diverse diseases, including neurodegeneration, cancer, diabetes, and inflammation. This review discusses the current understanding in both adaptive and apoptotic responses as well as the molecular mechanisms instigating apoptosis via IRE1 and PERK signaling. We also examine how IRE1 and PERK signaling may be differentially used during neurodegeneration arising in retinitis pigmentosa and prion infection.


Molecular Biology of the Cell | 2012

IRE1 directs proteasomal and lysosomal degradation of misfolded rhodopsin

Wei-Chieh Chiang; Carissa Messah; Jonathan H. Lin

IRE1 signaling robustly promoted P23H rhodopsin degradation by both proteasome and lysosome. When one degradation pathway was blocked, IRE1 signaling still promoted misfolded rhodopsin degradation using the remaining pathway. IRE1 signaling reduced levels of other misfolded rhodopsins, with lesser effects on misfolded CFTR.


Investigative Ophthalmology & Visual Science | 2012

Induction of Endoplasmic Reticulum Stress Genes, BiP and Chop, in Genetic and Environmental Models of Retinal Degeneration

Heike Kroeger; Carissa Messah; Kelly Ahern; Jason Gee; Victory Joseph; Michael T. Matthes; Douglas Yasumura; Marina S. Gorbatyuk; Wei-Chieh Chiang; Matthew M. LaVail; Jonathan H. Lin

PURPOSE Endoplasmic reticulum (ER) stress has been observed in animal models of retinitis pigmentosa expressing P23H rhodopsin. We compared levels of tightly induced ER stress genes, Binding of immunoglobulin protein (BiP) and CCAAT/enhancer-binding protein homologous protein (Chop), in seven additional models of retinal degeneration arising from genetic or environmental causes. METHODS Retinas from transgenic S334ter rhodopsin (lines 3, 4, and 5) and Royal College of Surgeons (RCS and RCS-p+) rats from postnatal (P) days 10 to 120 were analyzed. In a constant light (CL) model of retinal degeneration, BALB/c mice were exposed to 15,000 lux of CL for 0 to 8 hours. Retinal tissues from three to eight animals per experimental condition were collected for histologic and molecular analyses. RESULTS S334ter animals revealed significant increases in BiP, S334ter-3 (3.3× at P15), S334ter-4 (4× at P60), and S334ter-5 (2.2× at P90), and Chop, S334ter-3 (1.3× at P15), S334ter-4 (1.5× at P30), and S334ter-5 (no change), compared with controls. P23H-3 rats showed significant increase of BiP at P60 (2.3×) and Chop (1.6×). RCS and RCS-p+ rats showed significant increases in BiP at P60 (2.4×) and P20 (1.8×), respectively, but no statistically significant changes in Chop. BALB/c mice showed increases in BiP (1.5×) and Chop (1.3×) after 4 hours of CL. Increased levels of these ER stress markers correlated with photoreceptor cell loss. CONCLUSIONS Our study reveals surprising increases in BiP and to a lesser degree Chop in retinal degenerations arising from diverse causes. We propose that manipulation of ER stress responses may be helpful in treating many environmental and heritable forms of retinal degeneration.


Investigative Ophthalmology & Visual Science | 2012

Selective Activation of ATF6 and PERK Endoplasmic Reticulum Stress Signaling Pathways Prevent Mutant Rhodopsin Accumulation

Wei-Chieh Chiang; Nobuhiko Hiramatsu; Carissa Messah; Heike Kroeger; Jonathan H. Lin

PURPOSE Many rhodopsin mutations that cause retinitis pigmentosa produce misfolded rhodopsin proteins that are retained within the endoplasmic reticulum (ER) and cause photoreceptor cell death. Activating transcription factor 6 (ATF6) and protein kinase RNA-like endoplasmic reticulum kinase (PERK) control intracellular signaling pathways that maintain ER homeostasis. The aim of this study was to investigate how ATF6 and PERK signaling affected misfolded rhodopsin in cells, which could identify new molecular therapies to treat retinal diseases associated with ER protein misfolding. METHODS To examine the effect of ATF6 on rhodopsin, wild-type (WT) or mutant rhodopsins were expressed in cells expressing inducible human ATF6f, the transcriptional activator domain of ATF6. Induction of ATF6f synthesis rapidly activated downstream genes. To examine PERKs effect on rhodopsin, WT or mutant rhodopsins were expressed in cells expressing a genetically altered PERK protein, Fv2E-PERK. Addition of the dimerizing molecule (AP20187) rapidly activated Fv2E-PERK and downstream genes. By use of these strategies, it was examined how selective ATF6 or PERK signaling affected the fate of WT and mutant rhodopsins. RESULTS ATF6 significantly reduced T17M, P23H, Y178C, C185R, D190G, K296E, and S334ter rhodopsin protein levels in the cells with minimal effects on monomeric WT rhodopsin protein levels. By contrast, the PERK pathway reduced both levels of WT, mutant rhodopsins, and many other proteins in the cell. CONCLUSIONS This study indicates that selectively activating ATF6 or PERK prevents mutant rhodopsin from accumulating in cells. ATF6 signaling may be especially useful in treating retinal degenerative diseases arising from rhodopsin misfolding by preferentially clearing mutant rhodopsin and abnormal rhodopsin aggregates.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Achromatopsia mutations target sequential steps of ATF6 activation

Wei-Chieh Chiang; Priscilla Chan; Bernd Wissinger; Ajoy Vincent; Anna Skorczyk-Werner; Maciej R. Krawczynski; Randal J. Kaufman; Stephen H. Tsang; Elise Héon; Susanne Kohl; Jonathan H. Lin

Significance The unfolded protein response regulator activating transcription factor 6 (ATF6) was recently identified as a novel genetic cause of the cone photoreceptor disease achromatopsia. ATF6 upregulates genes that help cells cope with endoplasmic reticulum stress. We identified the pathomechanisms of all ATF6 achromatopsia mutations. Class 1 ATF6 mutants show impaired endoplasmic reticulum (ER)-to-Golgi trafficking and diminished production of the transcriptional activator fragment. Class 2 mutants encode the intact ATF6 transcriptional activator domain with full activity. Class 3 mutants have defective basic leucine zipper (bZIP) domains with abrogated function. Patient fibroblasts show increased apoptosis after ER stress. Our findings reveal that human ATF6 mutations interrupt distinct steps of ATF6 activation. ER stress-associated damage may underlie the pathology of achromatopsia arising from ATF6. Achromatopsia is an autosomal recessive disorder characterized by cone photoreceptor dysfunction. We recently identified activating transcription factor 6 (ATF6) as a genetic cause of achromatopsia. ATF6 is a key regulator of the unfolded protein response. In response to endoplasmic reticulum (ER) stress, ATF6 migrates from the ER to Golgi to undergo regulated intramembrane proteolysis to release a cytosolic domain containing a basic leucine zipper (bZIP) transcriptional activator. The cleaved ATF6 fragment migrates to the nucleus to transcriptionally up-regulate protein-folding enzymes and chaperones. ATF6 mutations in patients with achromatopsia include missense, nonsense, splice site, and single-nucleotide deletion or duplication changes found across the entire gene. Here, we comprehensively tested the function of achromatopsia-associated ATF6 mutations and found that they group into three distinct molecular pathomechanisms: class 1 ATF6 mutants show impaired ER-to-Golgi trafficking and diminished regulated intramembrane proteolysis and transcriptional activity; class 2 ATF6 mutants bear the entire ATF6 cytosolic domain with fully intact transcriptional activity and constitutive induction of downstream target genes, even in the absence of ER stress; and class 3 ATF6 mutants have complete loss of transcriptional activity because of absent or defective bZIP domains. Primary fibroblasts from patients with class 1 or class 3 ATF6 mutations show increased cell death in response to ER stress. Our findings reveal that human ATF6 mutations interrupt distinct sequential steps of the ATF6 activation mechanism. We suggest that increased susceptibility to ER stress-induced damage during retinal development underlies the pathology of achromatopsia in patients with ATF6 mutations.


Brain Research | 2016

Endoplasmic reticulum stress in human photoreceptor diseases

Priscilla Chan; Julia Stolz; Susanne Kohl; Wei-Chieh Chiang; Jonathan H. Lin

Photoreceptors are specialized sensory neurons essential for light detection in the human eye. Photoreceptor cell dysfunction and death cause vision loss in many eye diseases such as retinitis pigmentosa and achromatopsia. Endoplasmic reticulum (ER) stress and Unfolded Protein Response (UPR) signaling have been implicated in the development and pathology of heritable forms of retinitis pigmentosa and achromatopsia. We review the role of ER stress and UPR in retinitis pigmentosa arising from misfolded rhodopsins (RHO) and in achromatopsia arising from genetic mutations in Activating Transcription Factor 6 (ATF6). This article is part of a Special Issue entitled SI:ER stress.


Journal of Virology | 2014

De Novo Prion Aggregates Trigger Autophagy in Skeletal Muscle

Shivanjali Joshi-Barr; Cyrus Bett; Wei-Chieh Chiang; Margarita Trejo; Hans H. Goebel; Beata Sikorska; Pawel P. Liberski; Alex J. Raeber; Jonathan H. Lin; Eliezer Masliah; Christina J. Sigurdson

ABSTRACT In certain sporadic, familial, and infectious prion diseases, the prion protein misfolds and aggregates in skeletal muscle in addition to the brain and spinal cord. In myocytes, prion aggregates accumulate intracellularly, yet little is known about clearance pathways. Here we investigated the clearance of prion aggregates in muscle of transgenic mice that develop prion disease de novo. In addition to neurodegeneration, aged mice developed a degenerative myopathy, with scattered myocytes containing ubiquitinated, intracellular prion inclusions that were adjacent to myocytes lacking inclusions. Myocytes also showed elevated levels of the endoplasmic reticulum chaperone Grp78/BiP, suggestive of impaired protein degradation and endoplasmic reticulum stress. Additionally, autophagy was induced, as indicated by increased levels of beclin-1 and LC3-II. In C2C12 myoblasts, inhibition of autophagosome maturation or lysosomal degradation led to enhanced prion aggregation, consistent with a role for autophagy in prion aggregate clearance. Taken together, these findings suggest that the induction of autophagy may be a central strategy for prion aggregate clearance in myocytes. IMPORTANCE In prion diseases, the prion protein misfolds and aggregates in the central nervous system and sometimes in other organs, including muscle, yet the cellular pathways of prion aggregate clearance are unclear. Here we investigated the clearance of prion aggregates in the muscle of a transgenic mouse model that develops profound muscle degeneration. We found that endoplasmic reticulum stress pathways were activated and that autophagy was induced. Blocking of autophagic degradation in cell culture models led to an accumulation of aggregated prion protein. Collectively, these findings suggest that autophagy has an instrumental role in prion protein clearance.


Investigative Ophthalmology & Visual Science | 2015

In Vivo Visualization of Endoplasmic Reticulum Stress in the Retina Using the ERAI Reporter Mouse

Marcel V. Alavi; Wei-Chieh Chiang; Heike Kroeger; Douglas Yasumura; Michael T. Matthes; Takao Iwawaki; Matthew M. LaVail; Douglas B. Gould; Jonathan H. Lin

PURPOSE Endoplasmic reticulum (ER) stress activates inositol requiring enzyme 1 (IRE1), a key regulator of the unfolded protein response. The ER stress activated indicator (ERAI) transgenic mouse expresses a yellow fluorescent GFP variant (Venus) when IRE1 is activated by ER stress. We tested whether ERAI mice would allow for real-time longitudinal studies of ER stress in living mouse eyes. METHODS We chemically and genetically induced ER stress, and qualitatively and quantitatively studied the Venus signal by fluorescence ophthalmoscopy. We determined retinal cell types that contribute to the signal by immunohistology, and we performed molecular and biochemical assays using whole retinal lysates to assess activity of the IRE1 pathway. RESULTS We found qualitative increase in vivo in fluorescence signal at sites of intravitreal tunicamycin injection in ERAI eyes, and quantitative increase in ERAI mice mated to RhoP23H mice expressing ER stress-inducing misfolded rhodopsin protein. As expected, we found that increased Venus signal arose primarily from photoreceptors in RhoP23H/+;ERAI mice. We found increased Xbp1S and XBP1s transcriptional target mRNA levels in RhoP23H/+;ERAI retinas compared to Rho+/+;ERAI retinas, and that Venus signal increased in ERAI retinas as a function of age. CONCLUSIONS Fluorescence ophthalmoscopy of ERAI mice enables in vivo visualization of retinas undergoing ER stress. ER stress activated indicator mice enable identification of individual retinal cells undergoing ER stress by immunohistochemistry. ER stress activated indicator mice show higher Venus signal at older ages, likely arising from amplification of basal retinal ER stress levels by GFPs inherent stability.


Advances in Experimental Medicine and Biology | 2012

Endoplasmic Reticulum-Associated Degradation (ERAD) of Misfolded Glycoproteins and Mutant P23H Rhodopsin in Photoreceptor Cells

Heike Kroeger; Wei-Chieh Chiang; Jonathan H. Lin

Membrane proteins, such as rhodopsin, often undergo N-linked glycosylation after translocation into the endoplasmic reticulum (ER). N-linked glycans are markers for correct protein folding, protein quality control, transport, and recognition by the ER-associated degradation (ERAD) machinery. The ER contains many resident proteins that promote correct folding of newly synthesized proteins and prevent inappropriate aggregation of protein-folding intermediates. The quality control mechanisms of the ER guarantee that only correctly folded proteins exit the ER and progress through the secretory pathway. Here, we review the ERAD pathway for glycoproteins and discuss recent reports linking ERAD to the development of retinitis pigmentosa arising from misfolded rhodopsin.

Collaboration


Dive into the Wei-Chieh Chiang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Heike Kroeger

University of California

View shared research outputs
Top Co-Authors

Avatar

Carissa Messah

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Susanne Kohl

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge