Network


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

Hotspot


Dive into the research topics where Zhonggang Hou is active.

Publication


Featured researches published by Zhonggang Hou.


Nature Methods | 2011

Chemically defined conditions for human iPSC derivation and culture

Guokai Chen; Daniel R. Gulbranson; Zhonggang Hou; Jennifer M. Bolin; Victor Ruotti; Mitchell D Probasco; Kimberly Smuga-Otto; Sara E. Howden; Nicole R. Diol; Nicholas E. Propson; Ryan Wagner; Garrett O. Lee; Jessica Antosiewicz-Bourget; Joyce Teng; James A. Thomson

We re-examine the individual components for human embryonic stem cell (ESC) and induced pluripotent stem cell (iPSC) culture and formulate a cell culture system in which all protein reagents for liquid media, attachment surfaces and splitting are chemically defined. A major improvement is the lack of a serum albumin component, as variations in either animal- or human-sourced albumin batches have previously plagued human ESC and iPSC culture with inconsistencies. Using this new medium (E8) and vitronectin-coated surfaces, we demonstrate improved derivation efficiencies of vector-free human iPSCs with an episomal approach. This simplified E8 medium should facilitate both the research use and clinical applications of human ESCs and iPSCs and their derivatives, and should be applicable to other reprogramming methods.


Cell | 2013

Epigenomic Analysis of Multilineage Differentiation of Human Embryonic Stem Cells

Wei Xie; Matthew D. Schultz; Ryan Lister; Zhonggang Hou; Nisha Rajagopal; Pradipta Ray; John W. Whitaker; Shulan Tian; R. David Hawkins; Danny Leung; Hongbo Yang; Tao Wang; Ah Young Lee; Scott Swanson; Jiuchun Zhang; Yun Zhu; Audrey Kim; Joseph R. Nery; Mark A. Urich; Samantha Kuan; Chia An Yen; Sarit Klugman; Pengzhi Yu; Kran Suknuntha; Nicholas E. Propson; Huaming Chen; Lee Edsall; Ulrich Wagner; Yan Li; Zhen Ye

Epigenetic mechanisms have been proposed to play crucial roles in mammalian development, but their precise functions are only partially understood. To investigate epigenetic regulation of embryonic development, we differentiated human embryonic stem cells into mesendoderm, neural progenitor cells, trophoblast-like cells, and mesenchymal stem cells and systematically characterized DNA methylation, chromatin modifications, and the transcriptome in each lineage. We found that promoters that are active in early developmental stages tend to be CG rich and mainly engage H3K27me3 upon silencing in nonexpressing lineages. By contrast, promoters for genes expressed preferentially at later stages are often CG poor and primarily employ DNA methylation upon repression. Interestingly, the early developmental regulatory genes are often located in large genomic domains that are generally devoid of DNA methylation in most lineages, which we termed DNA methylation valleys (DMVs). Our results suggest that distinct epigenetic mechanisms regulate early and late stages of ES cell differentiation.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Efficient genome engineering in human pluripotent stem cells using Cas9 from Neisseria meningitidis

Zhonggang Hou; Yan Zhang; Nicholas E. Propson; Sara E. Howden; Li Fang Chu; Erik J. Sontheimer; James A. Thomson

Significance Genome engineering in human pluripotent stem cells holds great promise for biomedical research and regenerative medicine, but it is very challenging. Recently, an RNA-guided nuclease system called clustered, regularly interspaced, short palindromic repeats (CRISPR)/CRISPR-associated (Cas) has been applied to genome engineering, greatly increasing the efficiency of genome editing. Here, using a CRISPR-Cas system identified in Neisseria meningitidis, which is distinct from the commonly used Streptococcus pyogenes system, we demonstrate efficient genome engineering in human pluripotent stem cells. Our study could have a tremendous impact in regenerative medicine. Genome engineering in human pluripotent stem cells (hPSCs) holds great promise for biomedical research and regenerative medicine. Recently, an RNA-guided, DNA-cleaving interference pathway from bacteria [the type II clustered, regularly interspaced, short palindromic repeats (CRISPR)-CRISPR-associated (Cas) pathway] has been adapted for use in eukaryotic cells, greatly facilitating genome editing. Only two CRISPR-Cas systems (from Streptococcus pyogenes and Streptococcus thermophilus), each with their own distinct targeting requirements and limitations, have been developed for genome editing thus far. Furthermore, limited information exists about homology-directed repair (HDR)-mediated gene targeting using long donor DNA templates in hPSCs with these systems. Here, using a distinct CRISPR-Cas system from Neisseria meningitidis, we demonstrate efficient targeting of an endogenous gene in three hPSC lines using HDR. The Cas9 RNA-guided endonuclease from N. meningitidis (NmCas9) recognizes a 5′-NNNNGATT-3′ protospacer adjacent motif (PAM) different from those recognized by Cas9 proteins from S. pyogenes and S. thermophilus (SpCas9 and StCas9, respectively). Similar to SpCas9, NmCas9 is able to use a single-guide RNA (sgRNA) to direct its activity. Because of its distinct protospacer adjacent motif, the N. meningitidis CRISPR-Cas machinery increases the sequence contexts amenable to RNA-directed genome editing.


Cell Stem Cell | 2010

Actin-Myosin Contractility Is Responsible for the Reduced Viability of Dissociated Human Embryonic Stem Cells

Guokai Chen; Zhonggang Hou; Daniel R. Gulbranson; James A. Thomson

Human ESCs are the pluripotent precursor of the three embryonic germ layers. Human ESCs exhibit basal-apical polarity, junctional complexes, integrin-dependent matrix adhesion, and E-cadherin-dependent cell-cell adhesion, all characteristics shared by the epiblast epithelium of the intact mammalian embryo. After disruption of epithelial structures, programmed cell death is commonly observed. If individualized human ESCs are prevented from reattaching and forming colonies, their viability is significantly reduced. Here, we show that actin-myosin contraction is a critical effector of the cell death response to human ESC dissociation. Inhibition of myosin heavy chain ATPase, downregulation of myosin heavy chain, and downregulation of myosin light chain all increase survival and cloning efficiency of individualized human ESCs. ROCK inhibition decreases phosphorylation of myosin light chain, suggesting that inhibition of actin-myosin contraction is also the mechanism through which ROCK inhibitors increase cloning efficiency of human ESCs.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Human pluripotent stem cell-derived neural constructs for predicting neural toxicity

Michael P. Schwartz; Zhonggang Hou; Nicholas E. Propson; Jue Zhang; Collin J. Engstrom; Vítor Santos Costa; Peng Jiang; Bao Kim Nguyen; Jennifer M. Bolin; William T. Daly; Yu Wang; Ron Stewart; C. David Page; William L. Murphy; James A. Thomson

Significance Stem cell biology, tissue engineering, bioinformatics, and machine learning were combined to implement an in vitro human cellular model for developmental neurotoxicity screening. Human pluripotent stem cell-derived neural tissue constructs with vascular networks and microglia were produced with high sample uniformity by combining precursor cells on synthetic hydrogels using standard culture techniques. Machine learning was used to build a predictive model from changes in global gene expression for neural constructs exposed to 60 toxic and nontoxic training chemicals. The model correctly classified 9 of 10 additional chemicals in a blinded trial. This combined strategy demonstrates the value of human cell-based assays for predictive toxicology and should be useful for both drug and chemical safety assessment. Human pluripotent stem cell-based in vitro models that reflect human physiology have the potential to reduce the number of drug failures in clinical trials and offer a cost-effective approach for assessing chemical safety. Here, human embryonic stem (ES) cell-derived neural progenitor cells, endothelial cells, mesenchymal stem cells, and microglia/macrophage precursors were combined on chemically defined polyethylene glycol hydrogels and cultured in serum-free medium to model cellular interactions within the developing brain. The precursors self-assembled into 3D neural constructs with diverse neuronal and glial populations, interconnected vascular networks, and ramified microglia. Replicate constructs were reproducible by RNA sequencing (RNA-Seq) and expressed neurogenesis, vasculature development, and microglia genes. Linear support vector machines were used to construct a predictive model from RNA-Seq data for 240 neural constructs treated with 34 toxic and 26 nontoxic chemicals. The predictive model was evaluated using two standard hold-out testing methods: a nearly unbiased leave-one-out cross-validation for the 60 training compounds and an unbiased blinded trial using a single hold-out set of 10 additional chemicals. The linear support vector produced an estimate for future data of 0.91 in the cross-validation experiment and correctly classified 9 of 10 chemicals in the blinded trial.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Phosphorylation regulates human OCT4

Justin Brumbaugh; Zhonggang Hou; Jason D. Russell; Sara E. Howden; Pengzhi Yu; Aaron R. Ledvina; Joshua J. Coon; James A. Thomson

The transcription factor OCT4 is fundamental to maintaining pluripotency and self-renewal. To better understand protein-level regulation of OCT4, we applied liquid chromatography–MS to identify 14 localized sites of phosphorylation, 11 of which were previously unknown. Functional analysis of two sites, T234 and S235, suggested that phosphorylation within the homeobox region of OCT4 negatively regulates its activity by interrupting sequence-specific DNA binding. Mutating T234 and S235 to mimic constitutive phosphorylation at these sites reduces transcriptional activation from an OCT4-responsive reporter and decreases reprogramming efficiency. We also cataloged 144 unique phosphopeptides on known OCT4 interacting partners, including SOX2 and SALL4, that copurified during immunoprecipitation. These proteins were enriched for phosphorylation at motifs associated with ERK signaling. Likewise, OCT4 harbored several putative ERK phosphorylation sites. Kinase assays confirmed that ERK2 phosphorylated these sites in vitro, providing a direct link between ERK signaling and the transcriptional machinery that governs pluripotency.


Stem Cells | 2012

Thermal Stability of Fibroblast Growth Factor Protein Is a Determinant Factor in Regulating Self-Renewal, Differentiation, and Reprogramming in Human Pluripotent Stem Cells†‡§

Guokai Chen; Daniel R. Gulbranson; Pengzhi Yu; Zhonggang Hou; James A. Thomson

Fibroblast growth factor (FGF), transforming growth factor (TGF)/Nodal, and Insulin/insulin‐like growth factor (IGF) signaling pathways are sufficient to maintain human embryonic stem cells (ESCs) and induced pluripotent stem cells in a proliferative, undifferentiated state. Here, we show that only a few FGF family members (FGF2, FGF4, FGF6, and FGF9) are able to sustain strong extracellular‐signal‐regulated kinase (ERK) phosphorylation and NANOG expression levels in human ESCs. Surprisingly, FGF1, which is reported to target the same set of receptors as FGF2, fails to sustain ERK phosphorylation and NANOG expression under standard culture conditions. We find that the failure of FGF1 to sustain ES is due to thermal instability of the wild‐type protein, not receptor specificity, and that a mutated thermal‐stable FGF1 sustains human ESCs and supports both differentiation and reprogramming protocols. STEM CELLS 2012; 30:623–630


Genome Biology | 2016

Single-cell RNA-seq reveals novel regulators of human embryonic stem cell differentiation to definitive endoderm

Li-Fang Chu; Ning Leng; Jue Zhang; Zhonggang Hou; Daniel Mamott; David T. Vereide; Jeea Choi; Christina Kendziorski; Ron Stewart; James A. Thomson

BackgroundHuman pluripotent stem cells offer the best available model to study the underlying cellular and molecular mechanisms of human embryonic lineage specification. However, it is not fully understood how individual stem cells exit the pluripotent state and transition towards their respective progenitor states.ResultsHere, we analyze the transcriptomes of human embryonic stem cell-derived lineage-specific progenitors by single-cell RNA-sequencing (scRNA-seq). We identify a definitive endoderm (DE) transcriptomic signature that leads us to pinpoint a critical time window when DE differentiation is enhanced by hypoxia. The molecular mechanisms governing the emergence of DE are further examined by time course scRNA-seq experiments, employing two new statistical tools to identify stage-specific genes over time (SCPattern) and to reconstruct the differentiation trajectory from the pluripotent state through mesendoderm to DE (Wave-Crest). Importantly, presumptive DE cells can be detected during the transitory phase from Brachyury (T)+ mesendoderm toward a CXCR4+ DE state. Novel regulators are identified within this time window and are functionally validated on a screening platform with a T-2A-EGFP knock-in reporter engineered by CRISPR/Cas9. Through loss-of-function and gain-of-function experiments, we demonstrate that KLF8 plays a pivotal role modulating mesendoderm to DE differentiation.ConclusionsWe report the analysis of 1776 cells by scRNA-seq covering distinct human embryonic stem cell-derived progenitor states. By reconstructing a differentiation trajectory at single-cell resolution, novel regulators of the mesendoderm transition to DE are elucidated and validated. Our strategy of combining single-cell analysis and genetic approaches can be applied to uncover novel regulators governing cell fate decisions in a variety of systems.


PLOS Computational Biology | 2013

Integrated module and gene-specific regulatory inference implicates upstream signaling networks.

Sushmita Roy; Stephen Lagree; Zhonggang Hou; James A. Thomson; Ron Stewart; Audrey P. Gasch

Regulatory networks that control gene expression are important in diverse biological contexts including stress response and development. Each genes regulatory program is determined by module-level regulation (e.g. co-regulation via the same signaling system), as well as gene-specific determinants that can fine-tune expression. We present a novel approach, Modular regulatory network learning with per gene information (MERLIN), that infers regulatory programs for individual genes while probabilistically constraining these programs to reveal module-level organization of regulatory networks. Using edge-, regulator- and module-based comparisons of simulated networks of known ground truth, we find MERLIN reconstructs regulatory programs of individual genes as well or better than existing approaches of network reconstruction, while additionally identifying modular organization of the regulatory networks. We use MERLIN to dissect global transcriptional behavior in two biological contexts: yeast stress response and human embryonic stem cell differentiation. Regulatory modules inferred by MERLIN capture co-regulatory relationships between signaling proteins and downstream transcription factors thereby revealing the upstream signaling systems controlling transcriptional responses. The inferred networks are enriched for regulators with genetic or physical interactions, supporting the inference, and identify modules of functionally related genes bound by the same transcriptional regulators. Our method combines the strengths of per-gene and per-module methods to reveal new insights into transcriptional regulation in stress and development.


Acta Biomaterialia | 2016

Stable engineered vascular networks from human induced pluripotent stem cell-derived endothelial cells cultured in synthetic hydrogels.

Matthew R. Zanotelli; Hamisha Ardalani; Jue Zhang; Zhonggang Hou; Eric H. Nguyen; Scott Swanson; Bao Kim Nguyen; Jennifer M. Bolin; Angela L. Elwell; Lauren L. Bischel; Angela W. Xie; Ron Stewart; David J. Beebe; James A. Thomson; Michael P. Schwartz; William L. Murphy

UNLABELLED Here, we describe an in vitro strategy to model vascular morphogenesis where human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) are encapsulated in peptide-functionalized poly(ethylene glycol) (PEG) hydrogels, either on standard well plates or within a passive pumping polydimethylsiloxane (PDMS) tri-channel microfluidic device. PEG hydrogels permissive towards cellular remodeling were fabricated using thiol-ene photopolymerization to incorporate matrix metalloproteinase (MMP)-degradable crosslinks and CRGDS cell adhesion peptide. Time lapse microscopy, immunofluorescence imaging, and RNA sequencing (RNA-Seq) demonstrated that iPSC-ECs formed vascular networks through mechanisms that were consistent with in vivo vasculogenesis and angiogenesis when cultured in PEG hydrogels. Migrating iPSC-ECs condensed into clusters, elongated into tubules, and formed polygonal networks through sprouting. Genes upregulated for iPSC-ECs cultured in PEG hydrogels relative to control cells on tissue culture polystyrene (TCP) surfaces included adhesion, matrix remodeling, and Notch signaling pathway genes relevant to in vivo vascular development. Vascular networks with lumens were stable for at least 14days when iPSC-ECs were encapsulated in PEG hydrogels that were polymerized within the central channel of the microfluidic device. Therefore, iPSC-ECs cultured in peptide-functionalized PEG hydrogels offer a defined platform for investigating vascular morphogenesis in vitro using both standard and microfluidic formats. STATEMENT OF SIGNIFICANCE Human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) cultured in synthetic hydrogels self-assemble into capillary networks through mechanisms consistent with in vivo vascular morphogenesis.

Collaboration


Dive into the Zhonggang Hou's collaboration.

Top Co-Authors

Avatar

Michael P. Schwartz

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

William L. Murphy

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Daniel R. Gulbranson

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Guokai Chen

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Sara E. Howden

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

C. David Page

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Hamisha Ardalani

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Hongbo Yang

University of California

View shared research outputs
Top Co-Authors

Avatar

Huaming Chen

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge