Network


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

Hotspot


Dive into the research topics where Xiaosong Huang is active.

Publication


Featured researches published by Xiaosong Huang.


Cell Research | 2011

Efficient human iPS cell derivation by a non-integrating plasmid from blood cells with unique epigenetic and gene expression signatures

Bin Kuan Chou; Prashant Mali; Xiaosong Huang; Zhaohui Ye; Sarah N. Dowey; Linda M. S. Resar; Chunlin Zou; Y. Alex Zhang; Jay Tong; Linzhao Cheng

To identify accessible and permissive human cell types for efficient derivation of induced pluripotent stem cells (iPSCs), we investigated epigenetic and gene expression signatures of multiple postnatal cell types such as fibroblasts and blood cells. Our analysis suggested that newborn cord blood (CB) and adult peripheral blood (PB) mononuclear cells (MNCs) display unique signatures that are closer to iPSCs and human embryonic stem cells (ESCs) than age-matched fibroblasts to iPSCs/ESCs, thus making blood MNCs an attractive cell choice for the generation of integration-free iPSCs. Using an improved EBNA1/OriP plasmid expressing 5 reprogramming factors, we demonstrated highly efficient reprogramming of briefly cultured blood MNCs. Within 14 days of one-time transfection by one plasmid, up to 1000 iPSC-like colonies per 2 million transfected CB MNCs were generated. The efficiency of deriving iPSCs from adult PB MNCs was approximately 50-fold lower, but could be enhanced by inclusion of a second EBNA1/OriP plasmid for transient expression of additional genes such as SV40 T antigen. The duration of obtaining bona fide iPSC colonies from adult PB MNCs was reduced to half (∼14 days) as compared to adult fibroblastic cells (28–30 days). More than 9 human iPSC lines derived from PB or CB blood cells are extensively characterized, including those from PB MNCs of an adult patient with sickle cell disease. They lack V(D)J DNA rearrangements and vector DNA after expansion for 10–12 passages. This facile method of generating integration-free human iPSCs from blood MNCs will accelerate their use in both research and future clinical applications.


Stem Cells | 2015

Production of gene-corrected adult beta globin protein in human erythrocytes differentiated from patient iPSCs after genome editing of the sickle point mutation

Xiaosong Huang; Ying Wang; Wei Yan; Cory Smith; Zhaohui Ye; Jing Wang; Yongxing Gao; Laurel Mendelsohn; Linzhao Cheng

Human induced pluripotent stem cells (iPSCs) and genome editing provide a precise way to generate gene‐corrected cells for disease modeling and cell therapies. Human iPSCs generated from sickle cell disease (SCD) patients have a homozygous missense point mutation in the HBB gene encoding adult β‐globin proteins, and are used as a model system to improve strategies of human gene therapy. We demonstrate that the CRISPR/Cas9 system designer nuclease is much more efficient in stimulating gene targeting of the endogenous HBB locus near the SCD point mutation in human iPSCs than zinc finger nucleases and TALENs. Using a specific guide RNA and Cas9, we readily corrected one allele of the SCD HBB gene in human iPSCs by homologous recombination with a donor DNA template containing the wild‐type HBB DNA and a selection cassette that was subsequently removed to avoid possible interference of HBB transcription and translation. We chose targeted iPSC clones that have one corrected and one disrupted SCD allele for erythroid differentiation assays, using an improved xeno‐free and feeder‐free culture condition we recently established. Erythrocytes from either the corrected or its parental (uncorrected) iPSC line were generated with similar efficiencies. Currently ∼6%–10% of these differentiated erythrocytes indeed lacked nuclei, characteristic of further matured erythrocytes called reticulocytes. We also detected the 16‐kDa β‐globin protein expressed from the corrected HBB allele in the erythrocytes differentiated from genome‐edited iPSCs. Our results represent a significant step toward the clinical applications of genome editing using patient‐derived iPSCs to generate disease‐free cells for cell and gene therapies. Stem Cells 2015;33:1470–1479


Nature Protocols | 2012

Generation of integration-free human induced pluripotent stem cells from postnatal blood mononuclear cells by plasmid vector expression

Sarah N. Dowey; Xiaosong Huang; Bin Kuan Chou; Zhaohui Ye; Linzhao Cheng

Several human postnatal somatic cell types have been successfully reprogrammed to induced pluripotent stem cells (iPSCs). Blood mononuclear cells (MNCs) offer several advantages compared with other cell types. They are easily isolated from umbilical cord blood (CB) or adult peripheral blood (PB), and can be used fresh or after freezing. A short culture allows for more efficient reprogramming, with iPSC colonies forming from blood MNCs in 14 d, compared with 28 d for age-matched fibroblastic cells. The advantages of briefly cultured blood MNCs may be due to favorable epigenetic profiles and gene expression patterns. Blood cells from adults, especially nonlymphoid cells that are replenished frequently from intermittently activated blood stem cells, are short-lived in vivo and may contain less somatic mutations than skin fibroblasts, which are more exposed to environmental mutagens over time. We describe here a detailed, validated protocol for effective generation of integration-free human iPSCs from blood MNCs by plasmid vectors.


Stem Cells and Development | 2012

Efficient Derivation and Genetic Modifications of Human Pluripotent Stem Cells on Engineered Human Feeder Cell Lines

Chunlin Zou; Bin Kuan Chou; Sarah N. Dowey; Kitman Tsang; Xiaosong Huang; Cyndi F. Liu; Cory Smith; Jonathan Yen; Prashant Mali; Yu Alex Zhang; Linzhao Cheng; Zhaohui Ye

Derivation of pluripotent stem cells (iPSCs) induced from somatic cell types and the subsequent genetic modifications of disease-specific or patient-specific iPSCs are crucial steps in their applications for disease modeling as well as future cell and gene therapies. Conventional procedures of these processes require co-culture with primary mouse embryonic fibroblasts (MEFs) to support self-renewal and clonal growth of human iPSCs as well as embryonic stem cells (ESCs). However, the variability of MEF quality affects the efficiencies of all these steps. Furthermore, animal sourced feeders may hinder the clinical applications of human stem cells. In order to overcome these hurdles, we established immortalized human feeder cell lines by stably expressing human telomerase reverse transcriptase, Wnt3a, and drug resistance genes in adult mesenchymal stem cells. Here, we show that these immortalized human feeders support efficient derivation of virus-free, integration-free human iPSCs and long-term expansion of human iPSCs and ESCs. Moreover, the drug-resistance feature of these feeders also supports nonviral gene transfer and expression at a high efficiency, mediated by piggyBac DNA transposition. Importantly, these human feeders exhibit superior ability over MEFs in supporting homologous recombination-mediated gene targeting in human iPSCs, allowing us to efficiently target a transgene into the AAVS1 safe harbor locus in recently derived integration-free iPSCs. Our results have great implications in disease modeling and translational applications of human iPSCs, as these engineered human cell lines provide a more efficient tool for genetic modifications and a safer alternative for supporting self-renewal of human iPSCs and ESCs.


Molecular Therapy | 2014

Extensive Ex Vivo Expansion of Functional Human Erythroid Precursors Established From Umbilical Cord Blood Cells by Defined Factors

Xiaosong Huang; Siddharth Shah; Jing Wang; Zhaohui Ye; Sarah N. Dowey; Kit Man Tsang; Laurel Mendelsohn; Gregory J. Kato; Thomas S. Kickler; Linzhao Cheng

There is a constant shortage of red blood cells (RBCs) from sufficiently matched donors for patients who need chronic transfusion. Ex vivo expansion and maturation of human erythroid precursors (erythroblasts) from the patients or optimally matched donors could represent a potential solution. Proliferating erythroblasts can be expanded from umbilical cord blood mononuclear cells (CB MNCs) ex vivo for 10(6)-10(7)-fold (in ~50 days) before proliferation arrest and reaching sufficient number for broad application. Here, we report that ectopic expression of three genetic factors (Sox2, c-Myc, and an shRNA against TP53 gene) associated with iPSC derivation enables CB-derived erythroblasts to undergo extended expansion (~10(68)-fold in ~12 months) in a serum-free culture condition without change of cell identity or function. These expanding erythroblasts maintain immature erythroblast phenotypes and morphology, a normal diploid karyotype and dependence on a specific combination of growth factors for proliferation throughout expansion period. When being switched to a terminal differentiation condition, these immortalized erythroblasts gradually exit cell cycle, decrease cell size, accumulate hemoglobin, condense nuclei and eventually give rise to enucleated hemoglobin-containing erythrocytes that can bind and release oxygen. Our result may ultimately lead to an alternative approach to generate unlimited numbers of RBCs for personalized transfusion medicine.


Stem Cells | 2014

Differential Sensitivity to JAK Inhibitory Drugs by Isogenic Human Erythroblasts and Hematopoietic Progenitors Generated from Patient‐Specific Induced Pluripotent Stem Cells

Zhaohui Ye; Cyndi F. Liu; Lucie Lanikova; Sarah N. Dowey; Chaoxia He; Xiaosong Huang; Robert A. Brodsky; Jerry L. Spivak; Josef T. Prchal; Linzhao Cheng

Disease‐specific induced pluripotent stem cells (iPSCs) provide an unprecedented opportunity to establish novel disease models and accelerate drug development using distinct tissue target cells generated from isogenic iPSC lines with and without disease‐causing mutations. To realize the potential of iPSCs in modeling acquired diseases which are usually heterogeneous, we have generated multiple iPSC lines including two lines that are JAK2‐wild‐type and four lines homozygous for JAK2‐V617F somatic mutation from a single polycythemia vera (PV) patient blood. In vitro differentiation of the same patient‐derived iPSC lines have demonstrated the differential contributions of their parental hematopoietic clones to the abnormal erythropoiesis including the formation of endogenous erythroid colonies. This iPSC approach thus may provide unique and valuable insights into the genetic events responsible for disease development. To examine the potential of iPSCs in drug testing, we generated isogenic hematopoietic progenitors and erythroblasts from the same iPSC lines derived from PV patients and normal donors. Their response to three clinical JAK inhibitors, INCB018424 (Ruxolitinib), TG101348 (SAR302503), and the more recent CYT387 was evaluated. All three drugs similarly inhibited erythropoiesis from normal and PV iPSC lines containing the wild‐type JAK2 genotype, as well as those containing a homozygous or heterozygous JAK2‐V617F activating mutation that showed increased erythropoiesis without a JAK inhibitor. However, the JAK inhibitors had less inhibitory effect on the self‐renewal of CD34+ hematopoietic progenitors. The iPSC‐mediated disease modeling thus underlies the ineffectiveness of the current JAK inhibitors and provides a modeling system to develop better targeted therapies for the JAK2 mutated hematopoiesis. Stem Cells 2014;32:269–278


Stem Cells Translational Medicine | 2014

Concise Review: Stem Cell-Based Approaches to Red Blood Cell Production for Transfusion

Siddharth Shah; Xiaosong Huang; Linzhao Cheng

Blood transfusion is a common procedure in modern medicine, and it is practiced throughout the world; however, many countries report a less than sufficient blood supply. Even in developed countries where the supply is currently adequate, projected demographics predict an insufficient supply as early as 2050. The blood supply is also strained during occasional widespread disasters and crises. Transfusion of blood components such as red blood cells (RBCs), platelets, or neutrophils is increasingly used from the same blood unit for multiple purposes and to reduce alloimmune responses. Even for RBCs and platelets lacking nuclei and many antigenic cell‐surface molecules, alloimmunity could occur, especially in patients with chronic transfusion requirements. Once alloimmunization occurs, such patients require RBCs from donors with a different blood group antigen combination, making it a challenge to find donors after every successive episode of alloimmunization. Alternative blood substitutes such as synthetic oxygen carriers have so far proven unsuccessful. In this review, we focus on current research and technologies that permit RBC production ex vivo from hematopoietic stem cells, pluripotent stem cells, and immortalized erythroid precursors.


Blood | 2011

Site-specific gene correction of a point mutation in human iPS cells derived from an adult patient with sickle cell disease

Jizhong Zou; Prashant Mali; Xiaosong Huang; Sarah N. Dowey; Linzhao Cheng


Blood | 2011

Distinct Induced Pluripotent Stem Cell Clones with Somatic Mutations Prepared From PV Patients

Zhaohui Ye; Lucie Piterkova; Cyndi F. Liu; Sarah N. Dowey; Bin-Kuan Chou; Xiaosong Huang; Jerry L. Spivak; Sabina Swierczek; Alison R. Moliterno; Josef T. Prchal; Linzhao Cheng


Archive | 2014

derived from an adult patient with sickle cell disease Site-specific gene correction of a point mutation in human iPS cells

Jizhong Zou; Prashant Mali; Xiaosong Huang; Sarah N. Dowey; Linzhao Cheng

Collaboration


Dive into the Xiaosong Huang's collaboration.

Top Co-Authors

Avatar

Linzhao Cheng

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Sarah N. Dowey

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Zhaohui Ye

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Prashant Mali

University of California

View shared research outputs
Top Co-Authors

Avatar

Bin Kuan Chou

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Siddharth Shah

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Cyndi F. Liu

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Bin-Kuan Chou

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Cory Smith

Johns Hopkins University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge