Network


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

Hotspot


Dive into the research topics where Haiwei Mou is active.

Publication


Featured researches published by Haiwei Mou.


Human Gene Therapy | 2015

Adenovirus-Mediated Somatic Genome Editing of Pten by CRISPR/Cas9 in Mouse Liver in Spite of Cas9-Specific Immune Responses.

Dan Wang; Haiwei Mou; Shaoyong Li; Yingxiang Li; Soren Hough; Karen Tran; Jia Li; Hao Yin; Daniel G. Anderson; Erik J. Sontheimer; Zhiping Weng; Guangping Gao; Wen Xue

CRISPR/Cas9 derived from the bacterial adaptive immunity pathway is a powerful tool for genome editing, but the safety profiles of in vivo delivered Cas9 (including host immune responses to the bacterial Cas9 protein) have not been comprehensively investigated in model organisms. Nonalcoholic steatohepatitis (NASH) is a prevalent human liver disease characterized by excessive fat accumulation in the liver. In this study, we used adenovirus (Ad) vector to deliver a Streptococcus pyogenes-derived Cas9 system (SpCas9) targeting Pten, a gene involved in NASH and a negative regulator of the PI3K-AKT pathway, in mouse liver. We found that the Ad vector mediated efficient Pten gene editing even in the presence of typical Ad vector-associated immunotoxicity in the liver. Four months after vector infusion, mice receiving the Pten gene-editing Ad vector showed massive hepatomegaly and features of NASH, consistent with the phenotypes following Cre-loxP-induced Pten deficiency in mouse liver. We also detected induction of humoral immunity against SpCas9 and the potential presence of an SpCas9-specific cellular immune response. Our findings provide a strategy to model human liver diseases in mice and highlight the importance considering Cas9-specific immune responses in future translational studies involving in vivo delivery of CRISPR/Cas9.


Genome Medicine | 2015

Precision cancer mouse models through genome editing with CRISPR-Cas9

Haiwei Mou; Zachary Kennedy; Daniel G. Anderson; Hao Yin; Wen Xue

The cancer genome is highly complex, with hundreds of point mutations, translocations, and chromosome gains and losses per tumor. To understand the effects of these alterations, precise models are needed. Traditional approaches to the construction of mouse models are time-consuming and laborious, requiring manipulation of embryonic stem cells and multiple steps. The recent development of the clustered regularly interspersed short palindromic repeats (CRISPR)-Cas9 system, a powerful genome-editing tool for efficient and precise genome engineering in cultured mammalian cells and animals, is transforming mouse-model generation. Here, we review how CRISPR-Cas9 has been used to create germline and somatic mouse models with point mutations, deletions and complex chromosomal rearrangements. We highlight the progress and challenges of such approaches, and how these models can be used to understand the evolution and progression of individual tumors and identify new strategies for cancer treatment. The generation of precision cancer mouse models through genome editing will provide a rapid avenue for functional cancer genomics and pave the way for precision cancer medicine.


Nature Biotechnology | 2017

Structure-guided chemical modification of guide RNA enables potent non-viral in vivo genome editing

Hao Yin; Chun-Qing Song; Sneha Suresh; Qiongqiong Wu; Stephen Walsh; Luke Hyunsik Rhym; Esther Mintzer; Mehmet Fatih Bolukbasi; Lihua Julie Zhu; Kevin J. Kauffman; Haiwei Mou; Alicia Oberholzer; Junmei Ding; Suet-Yan Kwan; Roman L. Bogorad; Timofei S. Zatsepin; Victor Koteliansky; Scot A. Wolfe; Wen Xue; Robert Langer; Daniel G. Anderson

Efficient genome editing with Cas9–sgRNA in vivo has required the use of viral delivery systems, which have limitations for clinical applications. Translational efforts to develop other RNA therapeutics have shown that judicious chemical modification of RNAs can improve therapeutic efficacy by reducing susceptibility to nuclease degradation. Guided by the structure of the Cas9–sgRNA complex, we identify regions of sgRNA that can be modified while maintaining or enhancing genome-editing activity, and we develop an optimal set of chemical modifications for in vivo applications. Using lipid nanoparticle formulations of these enhanced sgRNAs (e-sgRNA) and mRNA encoding Cas9, we show that a single intravenous injection into mice induces >80% editing of Pcsk9 in the liver. Serum Pcsk9 is reduced to undetectable levels, and cholesterol levels are significantly lowered about 35% to 40% in animals. This strategy may enable non-viral, Cas9-based genome editing in the liver in clinical settings.


Genome Biology | 2017

CRISPR/Cas9-mediated genome editing induces exon skipping by alternative splicing or exon deletion.

Haiwei Mou; Jordan L. Smith; Lingtao Peng; Hao Yin; Jill Moore; Xiao-Ou Zhang; Chun-Qing Song; Ankur Sheel; Qiongqiong Wu; Deniz M. Ozata; Yingxiang Li; Daniel G. Anderson; Charles P. Emerson; Erik J. Sontheimer; Melissa J. Moore; Zhiping Weng; Wen Xue

CRISPR is widely used to disrupt gene function by inducing small insertions and deletions. Here, we show that some single-guide RNAs (sgRNAs) can induce exon skipping or large genomic deletions that delete exons. For example, CRISPR-mediated editing of β-catenin exon 3, which encodes an autoinhibitory domain, induces partial skipping of the in-frame exon and nuclear accumulation of β-catenin. A single sgRNA can induce small insertions or deletions that partially alter splicing or unexpected larger deletions that remove exons. Exon skipping adds to the unexpected outcomes that must be accounted for, and perhaps taken advantage of, in CRISPR experiments.


Gastroenterology | 2017

Genome-Wide CRISPR Screen Identifies Regulators of Mitogen-Activated Protein Kinase as Suppressors of Liver Tumors in Mice

Chun-Qing Song; Yingxiang Li; Haiwei Mou; Jill Moore; Angela Park; Yotsawat Pomyen; Soren Hough; Zachary Kennedy; Andrew H. Fischer; Hao Yin; Daniel G. Anderson; Darryl Conte; Lars Zender; Xin Wei Wang; Snorri S. Thorgeirsson; Zhiping Weng; Wen Xue

BACKGROUND & AIMS It has been a challenge to identify liver tumor suppressors or oncogenes due to the genetic heterogeneity of these tumors. We performed a genome-wide screen to identify suppressors of liver tumor formation in mice, using CRISPR-mediated genome editing. METHODS We performed a genome-wide CRISPR/Cas9-based knockout screen of P53-null mouse embryonic liver progenitor cells that overexpressed MYC. We infected p53-/-;Myc;Cas9 hepatocytes with the mGeCKOa lentiviral library of 67,000 single-guide RNAs (sgRNAs), targeting 20,611 mouse genes, and transplanted the transduced cells subcutaneously into nude mice. Within 1 month, all the mice that received the sgRNA library developed subcutaneous tumors. We performed high-throughput sequencing of tumor DNA and identified sgRNAs increased at least 8-fold compared to the initial cell pool. To validate the top 10 candidate tumor suppressors from this screen, we collected data from patients with hepatocellular carcinoma (HCC) using the Cancer Genome Atlas and COSMIC databases. We used CRISPR to inactivate candidate tumor suppressor genes in p53-/-;Myc;Cas9 cells and transplanted them subcutaneously into nude mice; tumor formation was monitored and tumors were analyzed by histology and immunohistochemistry. Mice with liver-specific disruption of p53 were given hydrodynamic tail-vein injections of plasmids encoding Myc and sgRNA/Cas9 designed to disrupt candidate tumor suppressors; growth of tumors and metastases was monitored. We compared gene expression profiles of liver cells with vs without tumor suppressor gene disrupted by sgRNA/Cas9. Genes found to be up-regulated after tumor suppressor loss were examined in liver cancer cell lines; their expression was knocked down using small hairpin RNAs, and tumor growth was examined in nude mice. Effects of the MEK inhibitors AZD6244, U0126, and trametinib, or the multi-kinase inhibitor sorafenib, were examined in human and mouse HCC cell lines. RESULTS We identified 4 candidate liver tumor suppressor genes not previously associated with liver cancer (Nf1, Plxnb1, Flrt2, and B9d1). CRISPR-mediated knockout of Nf1, a negative regulator of RAS, accelerated liver tumor formation in mice. Loss of Nf1 or activation of RAS up-regulated the liver progenitor cell markers HMGA2 and SOX9. RAS pathway inhibitors suppressed the activation of the Hmga2 and Sox9 genes that resulted from loss of Nf1 or oncogenic activation of RAS. Knockdown of HMGA2 delayed formation of xenograft tumors from cells that expressed oncogenic RAS. In human HCCs, low levels of NF1 messenger RNA or high levels of HMGA2 messenger RNA were associated with shorter patient survival time. Liver cancer cells with inactivation of Plxnb1, Flrt2, and B9d1 formed more tumors in mice and had increased levels of mitogen-activated protein kinase phosphorylation. CONCLUSIONS Using a CRISPR-based strategy, we identified Nf1, Plxnb1, Flrt2, and B9d1 as suppressors of liver tumor formation. We validated the observation that RAS signaling, via mitogen-activated protein kinase, contributes to formation of liver tumors in mice. We associated decreased levels of NF1 and increased levels of its downstream protein HMGA2 with survival times of patients with HCC. Strategies to inhibit or reduce HMGA2 might be developed to treat patients with liver cancer.


Cancer Cell | 2017

Oncogenic Activation of the RNA Binding Protein NELFE and MYC Signaling in Hepatocellular Carcinoma

Hien T. Dang; Atsushi Takai; Marshonna Forgues; Yotsowat Pomyen; Haiwei Mou; Wen Xue; Debashish Ray; Kevin C.H. Ha; Quaid Morris; Timothy R. Hughes; Xin Wei Wang

Global transcriptomic imbalance is a ubiquitous feature associated with cancer, including hepatocellular carcinoma (HCC). Analyses of 1,225 clinical HCC samples revealed that a large numbers of RNA binding proteins (RBPs) are dysregulated and that RBP dysregulation is associated with poor prognosis. We further identified that oncogenic activation of a top candidate RBP, negative elongation factor E (NELFE), via somatic copy-number alterations enhanced MYC signaling and promoted HCC progression. Interestingly, NELFE induces a unique tumor transcriptome by selectively regulating MYC-associated genes. Thus, our results revealed NELFE as an oncogenic protein that may contribute to transcriptome imbalance in HCC through the regulation of MYC signaling.


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

Genetic disruption of oncogenic Kras sensitizes lung cancer cells to Fas receptor-mediated apoptosis

Haiwei Mou; Jill Moore; Sunil K. Malonia; Yingxiang Li; Deniz M. Ozata; Soren Hough; Chun-Qing Song; Jordan L. Smith; Andrew H. Fischer; Zhiping Weng; Michael R. Green; Wen Xue

Significance Oncogenic KRAS underlies 30–90% of lung, colon, and pancreatic cancers, but despite more than 30 y of research, clinical inhibitors of KRAS—and potential resistance mechanisms—remain elusive. Using CRISPR-mediated genome editing of oncogenic Kras, we show that some lung cancer cells can survive Kras knockout, indicating the existence of mechanisms that allow tumors to escape Kras oncogene addiction. We identify genes highly expressed in Kras knockout cells, including the Fas receptor gene. Antibodies that activate Fas receptor selectively induced apoptosis in Kras-independent lung cancer cells, suggesting a potential strategy for combinatorial therapies against Kras-driven tumors. These findings have direct translational implications for the treatment of lung cancer and other KRAS mutant cancer types. Genetic lesions that activate KRAS account for ∼30% of the 1.6 million annual cases of lung cancer. Despite clinical need, KRAS is still undruggable using traditional small-molecule drugs/inhibitors. When oncogenic Kras is suppressed by RNA interference, tumors initially regress but eventually recur and proliferate despite suppression of Kras. Here, we show that tumor cells can survive knockout of oncogenic Kras, indicating the existence of Kras-independent survival pathways. Thus, even if clinical KRAS inhibitors were available, resistance would remain an obstacle to treatment. Kras-independent cancer cells exhibit decreased colony formation in vitro but retain the ability to form tumors in mice. Comparing the transcriptomes of oncogenic Kras cells and Kras knockout cells, we identified 603 genes that were specifically up-regulated in Kras knockout cells, including the Fas gene, which encodes a cell surface death receptor involved in physiological regulation of apoptosis. Antibodies recognizing Fas receptor efficiently induced apoptosis of Kras knockout cells but not oncogenic Kras-expressing cells. Increased Fas expression in Kras knockout cells was attributed to decreased association of repressive epigenetic marks at the Fas promoter. Concordant with this observation, treating oncogenic Kras cells with histone deacetylase inhibitor and Fas-activating antibody efficiently induced apoptosis, thus bypassing the need to inhibit Kras. Our results suggest that activation of Fas could be exploited as an Achilles’ heel in tumors initiated by oncogenic Kras.


PLOS Genetics | 2018

The temporal landscape of recursive splicing during Pol II transcription elongation in human cells

Xiao-Ou Zhang; Yu Fu; Haiwei Mou; Wen Xue; Zhiping Weng

Recursive splicing (RS) is an evolutionarily conserved process of removing long introns via multiple steps of splicing. It was first discovered in Drosophila and recently proven to occur also in humans. The detailed mechanism of recursive splicing is not well understood, in particular, whether it is kinetically coupled with transcription. To investigate the dynamic process that underlies recursive splicing, we systematically characterized 342 RS sites in three human cell types using published time-series data that monitored synchronized Pol II elongation and nascent RNA production with 4-thiouridine labeling. We found that half of the RS events occurred post-transcriptionally with long delays. For at least 18–47% RS introns, we detected RS junction reads only after detecting canonical splicing junction reads, supporting the notion that these introns were removed by both recursive splicing and canonical splicing. Furthermore, the choice of which splicing mechanism was used showed cell type specificity. Our results suggest that recursive splicing supplements, rather than replaces, canonical splicing for removing long introns.


Nature Biotechnology | 2018

Cas9-mediated allelic exchange repairs compound heterozygous recessive mutations in mice

Dan Wang; Jia Li; Chun-Qing Song; Karen Tran; Haiwei Mou; Pei-Hsuan Wu; Phillip W.L. Tai; Craig A Mendonca; Lingzhi Ren; Blake Y. Wang; Qin Su; Dominic J. Gessler; Phillip D. Zamore; Wen Xue; Guangping Gao

We report a genome-editing strategy to correct compound heterozygous mutations, a common genotype in patients with recessive genetic disorders. Adeno-associated viral vector delivery of Cas9 and guide RNA induces allelic exchange and rescues the disease phenotype in mouse models of hereditary tyrosinemia type I and mucopolysaccharidosis type I. This approach recombines non-mutated genetic information present in two heterozygous alleles into one functional allele without using donor DNA templates.We report a genome-editing strategy to correct compound heterozygous mutations, a common genotype in patients with recessive genetic disorders. Adeno-associated viral vector delivery of Cas9 and guide RNA induces allelic exchange and rescues the disease phenotype in mouse models of hereditary tyrosinemia type I and mucopolysaccharidosis type I. This approach recombines non-mutated genetic information present in two heterozygous alleles into one functional allele without using donor DNA templates.


Molecular Therapy | 2016

733. Somatically Repairing Compound Heterozygous Recessive Mutations by Chromosomal Cut-and-Paste for In Vivo Gene Therapy

Dan Wang; Haiwei Mou; Karen Tran; Jia Li; Blake Y. Wang; Dominic J. Gessler; Phillip W.L. Tai; Qin Su; Wen Xue; Guangping Gao

Patients affected by monogenic recessive genetic disorders often carry two different mutated alleles of the same gene, which is known as compound heterozygous. Theoretically, exchanging the genetic material between the two mutated alleles will reconstitute a mutation-free allele that can be therapeutic (FigureFigure). We hypothesized that generating DNA double-stranded breaks at the same location on both mutant alleles can induce allele exchange, reconstitute a mutation-free allele, and therefore yield therapeutic benefit. We first tested this hypothesis in a targeted knock-in mouse model that carries GFPN-term-intron-tdTomatoC-term and tdTomatoN-term-intron-GFPC-term expression cassettes, respectively, at the same genomic location on each copy of Chr 11. Therefore, allele exchange at the intronic region will reconstitute the full-length GFP and tdTomato, serving as a reporter system. We injected recombinant AAV (rAAV) vectors expressing SpCas9 and sgRNA targeting the intron into adult mice by tail vein injection. Five weeks later, we observed GFP and tdTomato fluorescence in cryosections of peripheral tissues including liver and heart, whereas there was no such fluorescence observed in the tissue samples from untreated mice, demonstrating that allele exchange occurred, and that the reconstituted alleles yielded protein expression. Furthermore, we generated mice that carry two different mutations of the Aspa gene as a compound heterozygous mouse model of Canavan disease. We treated these mice with rAAV vectors expressing SpCas9 and sgRNA targeting an intron between the two mutation sites. Three weeks after treatment, we detected reconstituted, mutation-free Aspa DNA sequence by allele-specific PCR and single-molecule, high-throughput DNA sequencing in the liver. The reconstituted Aspa allele carried insertion at the predicted SpCas9 cleavage site, indicating that the DNA allele exchange was mediated by the non-homologous end joining DNA repair pathway. We also observed allele exchange in mouse liver using the SaCas9 system. Evaluation of the therapeutic benefit following Cas9/sgRNA-mediated allele exchange in various compound heterozygous mouse models and patient cell lines is underway. The gene repairing strategy described here is a novel approach to tackling a broad range of autosomal recessive genetic disorders.View Large Image | Download PowerPoint Slide

Collaboration


Dive into the Haiwei Mou's collaboration.

Top Co-Authors

Avatar

Wen Xue

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Daniel G. Anderson

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Hao Yin

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Zhiping Weng

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chun-Qing Song

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Soren Hough

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Dan Wang

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Guangping Gao

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Jia Li

University of Massachusetts Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge