Network


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

Hotspot


Dive into the research topics where Shang Hsun Yang is active.

Publication


Featured researches published by Shang Hsun Yang.


Nature | 2008

Towards a transgenic model of Huntington’s disease in a non-human primate

Shang Hsun Yang; Pei-Hsun Cheng; Heather Banta; Karolina Piotrowska-Nitsche; Jin Jing Yang; Eric C.H. Cheng; Brooke R. Snyder; Katherine Larkin; Jun Liu; Jack Orkin; Zhi Hui Fang; Yoland Smith; Jocelyne Bachevalier; Stuart M. Zola; Shihua Li; Xiao-Jiang Li; Anthony W.S. Chan

Non-human primates are valuable for modelling human disorders and for developing therapeutic strategies; however, little work has been reported in establishing transgenic non-human primate models of human diseases. Huntington’s disease (HD) is an autosomal dominant neurodegenerative disorder characterized by motor impairment, cognitive deterioration and psychiatric disturbances followed by death within 10–15 years of the onset of the symptoms. HD is caused by the expansion of cytosine-adenine-guanine (CAG, translated into glutamine) trinucleotide repeats in the first exon of the human huntingtin (HTT) gene. Mutant HTT with expanded polyglutamine (polyQ) is widely expressed in the brain and peripheral tissues, but causes selective neurodegeneration that is most prominent in the striatum and cortex of the brain. Although rodent models of HD have been developed, these models do not satisfactorily parallel the brain changes and behavioural features observed in HD patients. Because of the close physiological, neurological and genetic similarities between humans and higher primates, monkeys can serve as very useful models for understanding human physiology and diseases. Here we report our progress in developing a transgenic model of HD in a rhesus macaque that expresses polyglutamine-expanded HTT. Hallmark features of HD, including nuclear inclusions and neuropil aggregates, were observed in the brains of the HD transgenic monkeys. Additionally, the transgenic monkeys showed important clinical features of HD, including dystonia and chorea. A transgenic HD monkey model may open the way to understanding the underlying biology of HD better, and to the development of potential therapies. Moreover, our data suggest that it will be feasible to generate valuable non-human primate models of HD and possibly other human genetic diseases.


Human Molecular Genetics | 2008

Accumulation of N-terminal mutant huntingtin in mouse and monkey models implicated as a pathogenic mechanism in Huntington's disease

Chuan En Wang; Suzanne Tydlacka; Adam L. Orr; Shang Hsun Yang; Rona K. Graham; Michael R. Hayden; Shihua Li; Anthony W.S. Chan; Xiao-Jiang Li

A number of mouse models expressing mutant huntingtin (htt) with an expanded polyglutamine (polyQ) domain are useful for studying the pathogenesis of Huntingtons disease (HD) and identifying appropriate therapies. However, these models exhibit neurological phenotypes that differ in their severity and nature. Understanding how transgenic htt leads to variable neuropathology in animal models would shed light on the pathogenesis of HD and help us to choose HD models for investigation. By comparing the expression of mutant htt at the transcriptional and protein levels in transgenic mice expressing N-terminal or full-length mutant htt, we found that the accumulation and aggregation of mutant htt in the brain is determined by htt context. HD mouse models demonstrating more severe phenotypes show earlier accumulation of N-terminal mutant htt fragments, which leads to the formation of htt aggregates that are primarily present in neuronal nuclei and processes, as well as glial cells. Similarly, transgenic monkeys expressing exon-1 htt with a 147-glutamine repeat (147Q) died early and showed abundant neuropil aggregates in swelling neuronal processes. Fractionation of HD150Q knock-in mice brains revealed an age-dependent accumulation of N-terminal mutant htt fragments in the nucleus and synaptosomes, and this accumulation was most pronounced in the striatum due to decreased proteasomal activity. Our findings suggest that the neuropathological phenotypes of HD stem largely from the accumulation of N-terminal mutant htt fragments and that this accumulation is determined by htt context and cell-type-dependent clearance of mutant htt.


Human Molecular Genetics | 2015

Functional disruption of the dystrophin gene in Rhesus Monkey Using CRISPR/Cas9

Yongchang Chen; Yinghui Zheng; Yu Kang; Weili Yang; Yuyu Niu; Xiangyu Guo; Zhuchi Tu; Chenyang Si; Hong Wang; Ruxiao Xing; Xiuqiong Pu; Shang Hsun Yang; Shihua Li; Weizhi Ji; Xiao-Jiang Li

CRISPR/Cas9 has been used to genetically modify genomes in a variety of species, including non-human primates. Unfortunately, this new technology does cause mosaic mutations, and we do not yet know whether such mutations can functionally disrupt the targeted gene or cause the pathology seen in human disease. Addressing these issues is necessary if we are to generate large animal models of human diseases using CRISPR/Cas9. Here we used CRISPR/Cas9 to target the monkey dystrophin gene to create mutations that lead to Duchenne muscular dystrophy (DMD), a recessive X-linked form of muscular dystrophy. Examination of the relative targeting rate revealed that Crispr/Cas9 targeting could lead to mosaic mutations in up to 87% of the dystrophin alleles in monkey muscle. Moreover, CRISPR/Cas9 induced mutations in both male and female monkeys, with the markedly depleted dystrophin and muscle degeneration seen in early DMD. Our findings indicate that CRISPR/Cas9 can efficiently generate monkey models of human diseases, regardless of inheritance patterns. The presence of degenerated muscle cells in newborn Cas9-targeted monkeys suggests that therapeutic interventions at the early disease stage may be effective at alleviating the myopathy.


Glia | 2015

MicroRNA-145 as one negative regulator of astrogliosis.

Chih Yen Wang; Shang Hsun Yang; Shun Fen Tzeng

Astrogliosis occurs at the lesion site within days to weeks after spinal cord injury (SCI) and involves the proliferation and hypertrophy of astrocytes, leading to glia scar formation. Changes in gene expression by deregulated microRNAs (miRNAs) are involved in the process of central nervous system neurodegeneration. Here, we report that mir‐145, a miRNA enriched in rat spinal neurons and astrocytes, was downregulated at 1 week and 1 month after SCI. Our in vitro studies using astrocytes prepared from neonatal spinal cord tissues indicated that potent inflammagen lipopolysaccharide downregulated mir‐145 expression in astrocytes, suggesting that SCI‐triggered inflammatory signaling pathways could play the inhibitory role in astrocytic mir‐145 expression. To induce overexpression of mir‐145 in astrocytes at the spinal cord lesion site, we developed a lentivirus‐mediated pre‐miRNA delivery system using the promoter of glial fibrillary acidic protein (GFAP), an astrocyte‐specific intermediate filament. The results indicated that astrocyte‐specific overexpression of mir‐145 reduced astrocytic cell density at the lesion border of the injured spinal cord. In parallel, overexpression of mir‐145 reduced the size of astrocytes and the number of related cell processes, as well as cell proliferation and migration. Through a luciferase reporter system, we found that GFAP and c‐myc were the two potential targets of mir‐145 in astrocytes. Together, the findings demonstrate the novel role of mir‐145 in the regulation of astrocytic dynamics, and reveal that the downregulation of mir‐145 in astrocytes is a critical factor inducing astrogliosis after SCI. GLIA 2015;63:194–205


American Journal of Human Genetics | 2013

miR-196a Ameliorates Phenotypes of Huntington Disease in Cell, Transgenic Mouse, and Induced Pluripotent Stem Cell Models

Pei Hsun Cheng; Chia Ling Li; Yu Fan Chang; Shaw Jeng Tsai; Yen Yu Lai; Anthony W.S. Chan; Chuan-Mu Chen; Shang Hsun Yang

Huntington disease (HD) is a dominantly inherited neurodegenerative disorder characterized by dysregulation of various genes. Recently, microRNAs (miRNAs) have been reported to be involved in this dysregulation, suggesting that manipulation of appropriate miRNA regulation may have a therapeutic benefit. Here, we report the beneficial effects of miR-196a (miR196a) on HD in cell, transgenic mouse models, and human induced pluripotent stem cells derived from one individual with HD (HD-iPSCs). In the in vitro results, a reduction of mutant HTT and pathological aggregates, accompanying the overexpression of miR-196a, was observed in HD models of human embryonic kidney cells and mouse neuroblastoma cells. In the in vivo model, HD transgenic mice overexpressing miR-196a revealed the suppression of mutant HTT in the brain and also showed improvements in neuropathological progression, such as decreases of nuclear, intranuclear, and neuropil aggregates and late-stage behavioral phenotypes. Most importantly, miR-196a also decreased HTT expression and pathological aggregates when HD-iPSCs were differentiated into the neuronal stage. Mechanisms of miR-196a in HD might be through the alteration of ubiquitin-proteasome systems, gliosis, cAMP response element-binding protein pathway, and several neuronal regulatory pathways in vivo. Taken together, these results show that manipulating miR-196a provides beneficial effects in HD, suggesting the potential therapeutical role of miR-196a in HD.


Reproduction | 2008

Development of single mouse blastomeres into blastocysts, outgrowths and the establishment of embryonic stem cells

Chanchao Lorthongpanich; Shang Hsun Yang; Karolina Piotrowska-Nitsche; Rangsun Parnpai; Anthony W.S. Chan

The recently developed technique of establishing embryonic stem (ES) cell lines from single blastomeres (BTMs) of early mouse and human embryos has created significant interest in this source of ES cells. However, sister BTMs of an early embryo might not have equal competence for the development of different lineages or the derivation of ES cells. Therefore, single BTMs from two- and four-cell embryos of outbred mice were individually placed in sequential cultures to enhance the formation of the inner cell mass (ICM) and the establishment of embryonic outgrowth. The outgrowths were then used for the derivation of ES cell lines. Based on the expression of ICM (Sox2) and trophectoderm (Cdx2) markers, it was determined that ICM marker was lacking in blastocysts derived from 12% of BTMs from two-cell stage and 20% from four-cell stage. Four ES cell lines (5.6%; 4/72) were established ater culture of single BTMs from two-cell embryos, and their pluripotency was demonstrated by their differentiation into neuronal cell types. Our results demonstrate that sister BTMs of an early embryo are not equally competent for ICM marker expression. However, we demonstrated the feasibility of establishing ES cells from a single BTM of outbred mice.


European Journal of Clinical Investigation | 2006

Stem cells in the lung parenchyma and prospects for lung injury therapy

Chih-Ching Yen; Shang Hsun Yang; Chia-Chi Lin; Chuan-Mu Chen

Until recently, it was thought that only embryonic stem cells were pluripotent and that adult stem cells were restricted in their differentiative and regenerative potential to become the tissues in which they reside. However, the discovery that adult stem cells in one tissue can contribute to the formation of other tissues, especially after injury or cell damage, implies that stem cells have developmental plasticity. For example, haematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs) from bone marrow can be used to regenerate diverse tissues at distant sites, including the lung. This article reviews the character of stem cells in the lung parenchyma and focuses on the potential uses of adult stem cells in research of lung injury and lung disease therapies.


Methods | 2009

Generation of transgenic monkeys with human inherited genetic disease

Anthony W.S. Chan; Shang Hsun Yang

Modeling human diseases using nonhuman primates including chimpanzee, rhesus, cynomolgus, marmoset and squirrel monkeys has been reported in the past decades. Due to the high similarity between nonhuman primates and humans, including genome constitution, cognitive behavioral functions, anatomical structure, metabolic, reproductive, and brain functions; nonhuman primates have played an important role in understanding physiological functions of the human body, clarifying the underlying mechanism of human diseases, and the development of novel treatments for human diseases. However, nonhuman primate research has been restricted to cognitive, behavioral, biochemical and pharmacological approaches of human diseases due to the limitation of gene transfer technology in nonhuman primates. The recent advancement in transgenic technology that has led to the generation of the first transgenic monkey in 2001 and a transgenic monkey model of Huntingtons disease (HD) in 2008 has changed that focus. The creation of transgenic HD monkeys that replicate key pathological features of human HD patients further suggests the crucial role of nonhuman primates in the future development of biomedicine. These successes have opened the door to genetic manipulation in nonhuman primates and a new era in modeling human inherited genetic disorders. We focused on the procedures in creating transgenic Huntingtons disease monkeys, but our work can be applied to transgenesis in other nonhuman primate species.


Human Molecular Genetics | 2015

Early Parkinson's disease symptoms in α-synuclein transgenic monkeys

Yuyu Niu; Xiangyu Guo; Yongchang Chen; Chuan En Wang; Jinquan Gao; Weili Yang; Yu Kang; Wei Si; Hong Wang; Shang Hsun Yang; Shihua Li; Weizhi Ji; Xiao-Jiang Li

Parkinsons disease (PD) is an age-dependent neurodegenerative disease that can be caused by genetic mutations in α-synuclein (α-syn) or duplication of wild-type α-syn; PD is characterized by the deposition of α-syn aggregates, indicating a gain of toxicity from accumulation of α-syn. Although the major neuropathologic feature of PD is the degeneration of dopaminergic (DA) neurons in the substantia nigra, non-motor symptoms including anxiety, cognitive defect and sleep disorder precede the onset of motor impairment, and many clinical symptoms of PD are not caused by degeneration of DA neurons. Non-human primate models of PD are important for revealing the early pathology in PD and identifying effective treatments. We established transgenic PD rhesus monkeys that express mutant α-syn (A53T). Six transgenic A53T monkeys were produced via lentiviral vector expressing A53T in fertilized monkey eggs and subsequent embryo transfer to surrogates. Transgenic A53T is expressed in the monkey brain and causes age-dependent non-motor symptoms, including cognitive defects and anxiety phenotype, without detectable sleeping disorders. The transgenic α-syn monkeys demonstrate the specific early symptoms caused by mutant α-syn and provide insight into treatment of early PD.


PLOS ONE | 2012

Differential differences in methylation status of putative imprinted genes among cloned swine genomes.

Chih Jie Shen; Winston T.K. Cheng; Shinn-Chih Wu; Hsiao Ling Chen; Tung Chou Tsai; Shang Hsun Yang; Chuan-Mu Chen

DNA methylation is a major epigenetic modification in the mammalian genome that regulates crucial aspects of gene function. Mammalian cloning by somatic cell nuclear transfer (SCNT) often results in gestational or neonatal failure with only a small proportion of manipulated embryos producing live births. Many of the embryos that survive to term later succumb to a variety of abnormalities that are likely due to inappropriate epigenetic reprogramming. Aberrant methylation patterns of imprinted genes in cloned cattle and mice have been elucidated, but few reports have analyzed the cloned pig genome. Four surviving cloned sows that were created by ear fibroblast nuclear transfer, each with a different life span and multiple organ defects, such as heart defects and bone growth delay, were used as epigenetic study materials. First, we identified four putative differential methylation regions (DMR) of imprinted genes in the wild-type pig genome, including two maternally imprinted loci (INS and IGF2) and two paternally imprinted loci (H19 and IGF2R). Aberrant DNA methylation, either hypermethylation or hypomethylation, commonly appeared in H19 (45% of imprinted loci hypermethylated vs. 30% hypomethylated), IGF2 (40% vs. 0%), INS (50% vs. 5%), and IGF2R (15% vs. 45%) in multiple tissues from these four cloned sows compared with wild-type pigs. Our data suggest that aberrant epigenetic modifications occur frequently in the genome of cloned swine. Even with successful production of cloned swine that avoid prenatal or postnatal death, the perturbation of methylation in imprinted genes still exists, which may be one of reason for their adult pathologies and short life. Understanding the aberrant pattern of gene imprinting would permit improvements in future cloning techniques.

Collaboration


Dive into the Shang Hsun Yang's collaboration.

Top Co-Authors

Avatar

Chuan-Mu Chen

National Chung Hsing University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hsiao Ling Chen

National Chung Hsing University

View shared research outputs
Top Co-Authors

Avatar

Pei-Hsun Cheng

Yerkes National Primate Research Center

View shared research outputs
Top Co-Authors

Avatar

Pei Hsun Cheng

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Yu Fan Chang

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chia Ling Li

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

H. Sunny Sun

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Shaw Jenq Tsai

National Cheng Kung University

View shared research outputs
Researchain Logo
Decentralizing Knowledge