Network


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

Hotspot


Dive into the research topics where Lily Xia is active.

Publication


Featured researches published by Lily Xia.


Stem Cells | 2007

Sarcoma derived from cultured mesenchymal stem cells

Jakub Tolar; Alma J. Nauta; Mark J. Osborn; Angela Mortari; Ron T. McElmurry; Scott Bell; Lily Xia; Ning Zhou; Megan Riddle; Tania M. Schroeder; Jennifer J. Westendorf; R. Scott McIvor; Pancras C.W. Hogendoorn; Karoly Szuhai; LeAnn Oseth; Betsy Hirsch; Stephen R. Yant; Mark A. Kay; Alexandra Peister; Darwin J. Prockop; Willem E. Fibbe; Bruce R. Blazar

To study the biodistribution of MSCs, we labeled adult murine C57BL/6 MSCs with firefly luciferase and DsRed2 fluorescent protein using nonviral Sleeping Beauty transposons and coinfused labeled MSCs with bone marrow into irradiated allogeneic recipients. Using in vivo whole‐body imaging, luciferase signals were shown to be increased between weeks 3 and 12. Unexpectedly, some mice with the highest luciferase signals died and all surviving mice developed foci of sarcoma in their lungs. Two mice also developed sarcomas in their extremities. Common cytogenetic abnormalities were identified in tumor cells isolated from different animals. Original MSC cultures not labeled with transposons, as well as independently isolated cultured MSCs, were found to be cytogenetically abnormal. Moreover, primary MSCs derived from the bone marrow of both BALB/c and C57BL/6 mice showed cytogenetic aberrations after several passages in vitro, showing that transformation was not a strain‐specific nor rare event. Clonal evolution was observed in vivo, suggesting that the critical transformation event(s) occurred before infusion. Mapping of the transposition insertion sites did not identify an obvious transposon‐related genetic abnormality, and p53 was not overexpressed. Infusion of MSC‐derived sarcoma cells resulted in malignant lesions in secondary recipients. This new sarcoma cell line, S1, is unique in having a cytogenetic profile similar to human sarcoma and contains bioluminescent and fluorescent genes, making it useful for investigations of cellular biodistribution and tumor response to therapy in vivo. More importantly, our study indicates that sarcoma can evolve from MSC cultures.


Molecular Therapy | 2013

TALEN-based gene correction for epidermolysis bullosa.

Mark J. Osborn; Colby G. Starker; Amber N. McElroy; Beau R. Webber; Megan Riddle; Lily Xia; Anthony P. Defeo; Richard Gabriel; Manfred Schmidt; Christof von Kalle; Daniel F. Carlson; Morgan L. Maeder; J. Keith Joung; John E. Wagner; Daniel F. Voytas; Bruce R. Blazar; Jakub Tolar

Recessive dystrophic epidermolysis bullosa (RDEB) is characterized by a functional deficit of type VII collagen protein due to gene defects in the type VII collagen gene (COL7A1). Gene augmentation therapies are promising, but run the risk of insertional mutagenesis. To abrogate this risk, we explored the possibility of using engineered transcription activator-like effector nucleases (TALEN) for precise genome editing. We report the ability of TALEN to induce site-specific double-stranded DNA breaks (DSBs) leading to homology-directed repair (HDR) from an exogenous donor template. This process resulted in COL7A1 gene mutation correction in primary fibroblasts that were subsequently reprogrammed into inducible pluripotent stem cells and showed normal protein expression and deposition in a teratoma-based skin model in vivo. Deep sequencing-based genome-wide screening established a safety profile showing on-target activity and three off-target (OT) loci that, importantly, were at least 10 kb from a coding sequence. This study provides proof-of-concept for TALEN-mediated in situ correction of an endogenous patient-specific gene mutation and used an unbiased screen for comprehensive TALEN target mapping that will cooperatively facilitate translational application.


Blood | 2008

Amelioration of epidermolysis bullosa by transfer of wild-type bone marrow cells

Jakub Tolar; Akemi Ishida-Yamamoto; Megan Riddle; Ron T. McElmurry; Mark J. Osborn; Lily Xia; Troy C. Lund; Catherine Slattery; Jouni Uitto; Angela M. Christiano; John E. Wagner; Bruce R. Blazar

The recessive dystrophic form of epidermolysis bullosa (RDEB) is a disorder of incurable skin fragility and blistering caused by mutations in the type VII collagen gene (Col7a1). The absence of type VII collagen production leads to the loss of adhesion at the basement membrane zone due to the absence of anchoring fibrils, which are composed of type VII collagen. We report that wild-type, congenic bone marrow cells homed to damaged skin, produced type VII collagen protein and anchoring fibrils, ameliorated skin fragility, and reduced lethality in the murine model of RDEB generated by targeted Col7a1 disruption. These data provide the first evidence that a population of marrow cells can correct the basement membrane zone defect found in mice with RDEB and offer a potentially valuable approach for treatment of human RDEB and other extracellular matrix disorders.


Journal of Investigative Dermatology | 2011

Induced Pluripotent Stem Cells from Individuals with Recessive Dystrophic Epidermolysis Bullosa

Jakub Tolar; Lily Xia; Megan Riddle; Christopher J. Lees; Cindy R. Eide; Ron T. McElmurry; Matthias Titeux; Mark J. Osborn; Troy C. Lund; Alain Hovnanian; John E. Wagner; Bruce R. Blazar

Recessive dystrophic epidermolysis bullosa (RDEB) is an inherited blistering skin disorder caused by mutations in the COL7A1 gene-encoding type VII collagen (Col7), the major component of anchoring fibrils at the dermal-epidermal junction. Individuals with RDEB develop painful blisters and mucosal erosions, and currently, there are no effective forms of therapy. Nevertheless, some advances in patient therapy are being made, and cell-based therapies with mesenchymal and hematopoietic cells have shown promise in early clinical trials. To establish a foundation for personalized, gene-corrected, patient-specific cell transfer, we generated induced pluripotent stem (iPS) cells from three subjects with RDEB (RDEB iPS cells). We found that Col7 was not required for stem cell renewal and that RDEB iPS cells could be differentiated into both hematopoietic and nonhematopoietic lineages. The specific epigenetic profile associated with de-differentiation of RDEB fibroblasts and keratinocytes into RDEB iPS cells was similar to that observed in wild-type (WT) iPS cells. Importantly, human WT and RDEB iPS cells differentiated in vivo into structures resembling the skin. Gene-corrected RDEB iPS cells expressed Col7. These data identify the potential of RDEB iPS cells to generate autologous hematopoietic grafts and skin cells with the inherent capacity to treat skin and mucosal erosions that typify this genodermatosis.


Blood | 2011

Hematopoietic differentiation of induced pluripotent stem cells from patients with mucopolysaccharidosis type I (Hurler syndrome)

Jakub Tolar; In-Hyun Park; Lily Xia; Christopher J. Lees; Brandon Peacock; Beau R. Webber; Ron T. McElmurry; Cindy R. Eide; Paul J. Orchard; Michael Kyba; Mark J. Osborn; Troy C. Lund; John E. Wagner; George Q. Daley; Bruce R. Blazar

Mucopolysaccharidosis type I (MPS IH; Hurler syndrome) is a congenital deficiency of α-L-iduronidase, leading to lysosomal storage of glycosaminoglycans that is ultimately fatal following an insidious onset after birth. Hematopoietic cell transplantation (HCT) is a life-saving measure in MPS IH. However, because a suitable hematopoietic donor is not found for everyone, because HCT is associated with significant morbidity and mortality, and because there is no known benefit of immune reaction between the host and the donor cells in MPS IH, gene-corrected autologous stem cells may be the ideal graft for HCT. Thus, we generated induced pluripotent stem cells from 2 patients with MPS IH (MPS-iPS cells). We found that α-L-iduronidase was not required for stem cell renewal, and that MPS-iPS cells showed lysosomal storage characteristic of MPS IH and could be differentiated to both hematopoietic and nonhematopoietic cells. The specific epigenetic profile associated with de-differentiation of MPS IH fibroblasts into MPS-iPS cells was maintained when MPS-iPS cells are gene-corrected with virally delivered α-L-iduronidase. These data underscore the potential of MPS-iPS cells to generate autologous hematopoietic grafts devoid of immunologic complications of allogeneic transplantation, as well as generating nonhematopoietic cells with the potential to treat anatomical sites not fully corrected with HCT.


Blood | 2009

Multipotent adult progenitor cells can suppress graft-versus-host disease via prostaglandin E2 synthesis and only if localized to sites of allopriming

Steven L. Highfill; Ryan M. Kelly; Matthew J. O'Shaughnessy; Qing Zhou; Lily Xia; Angela Panoskaltsis-Mortari; Patricia A. Taylor; Jakub Tolar; Bruce R. Blazar

Multipotent adult progenitor cells (MAPCs) are nonhematopoietic stem cells capable of giving rise to a broad range of tissue cells. As such, MAPCs hold promise for tissue injury repair after transplant. In vitro, MAPCs potently suppressed allogeneic T-cell activation and proliferation in a dose-dependent, cell contact-independent, and T-regulatory cell-independent manner. Suppression occurred primarily through prostaglandin E(2) synthesis in MAPCs, which resulted in decreased proinflammatory cytokine production. When given systemically, MAPCs did not home to sites of allopriming and did not suppress graft-versus-host disease (GVHD). To ensure that MAPCs would colocalize with donor T cells, MAPCs were injected directly into the spleen at bone marrow transplantation. MAPCs limited donor T-cell proliferation and GVHD-induced injury via prostaglandin E(2) synthesis in vivo. Moreover, MAPCs altered the balance away from positive and toward inhibitory costimulatory pathway expression in splenic T cells and antigen-presenting cells. These findings are the first to describe the immunosuppressive capacity and mechanism of MAPC-induced suppression of T-cell alloresponses and illustrate the requirement for MAPC colocalization to sites of initial donor T-cell activation for GVHD inhibition. Such data have implications for the use of allogeneic MAPCs and possibly other immunomodulatory nonhematopoietic stem cells for preventing GVHD in the clinic.


Journal of Investigative Dermatology | 2014

Patient-Specific Naturally Gene-Reverted Induced Pluripotent Stem Cells in Recessive Dystrophic Epidermolysis Bullosa

Jakub Tolar; John A. McGrath; Lily Xia; Megan Riddle; Christopher J. Lees; Cindy R. Eide; Douglas R. Keene; Lu Liu; Mark J. Osborn; Troy C. Lund; Bruce R. Blazar; John E. Wagner

Spontaneous reversion of disease-causing mutations has been observed in some genetic disorders. In our clinical observations of severe generalized recessive dystrophic epidermolysis bullosa (RDEB), a currently incurable blistering genodermatosis caused by loss-of-function mutations in COL7A1 that results in a deficit of type VII collagen (C7), we have observed patches of healthy-appearing skin on some individuals. When biopsied, this skin revealed somatic mosaicism resulting from the self-correction of C7 deficiency. We believe this source of cells could represent an opportunity for translational “natural” gene therapy. We show that revertant RDEB keratinocytes expressing functional C7 can be reprogrammed into induced pluripotent stem cells (iPSCs) and that self-corrected RDEB iPSCs can be induced to differentiate into either epidermal or hematopoietic cell populations. Our results give proof in principle that an inexhaustible supply of functional patient-specific revertant cells can be obtained—potentially relevant to local wound therapy and systemic hematopoietic cell transplantation. This technology may also avoid some of the major limitations of other cell therapy strategies, e.g., immune rejection and insertional mutagenesis, which are associated with viral- and nonviral- mediated gene therapy. We believe this approach should be the starting point for autologous cellular therapies using “natural” gene therapy in RDEB and other diseases.


Journal of Investigative Dermatology | 2013

Keratinocytes from Induced Pluripotent Stem Cells in Junctional Epidermolysis Bullosa

Jakub Tolar; Lily Xia; Christopher J. Lees; Megan Riddle; Amber N. McElroy; Douglas R. Keene; Troy C. Lund; Mark J. Osborn; M. Peter Marinkovich; Bruce R. Blazar; John E. Wagner

Keratinocytes and dermal fibroblasts express adhesive proteins that ensure the epidermis remains attached to the skin basement membrane and to the papillary dermis. Congenital deficiency of any of at least 15 such proteins results in a blistering condition, termed epidermolysis bullosa (EB)(Fine et al., 2008). The most severe form of EB is the Herlitz variant of junctional EB (JEB-H) caused by loss-of-function mutations in one of the three genes (LAMA3, LAMB3, and LAMC2) encoding one of three chains of the heterotrimeric protein laminin 332 (LM-332)(Kiritsi et al., 2011). LM-332 is secreted by keratinocytes and interacts with integrin receptors α3β1 and α6β4 to form focal adhesions and stable anchoring contacts in the dermalepidermal junction (DEJ). Children with this autosomal recessive genodermatosis develop generalized skin blistering; extensive mucosal erosions in the upper respiratory, gastrointestinal, and genitourinary tracts; infections; and, despite supportive measures, typically die within the first year of life. Even though it has been largely accepted that JEB-H is untreatable(Yuen et al., 2012), evidence from a gene therapy trial for the less severe (non-Herlitz) form of JEB(Mavilio et al., 2006), and from LAMB3 gene correction of human JEB-H cells –(Robbins et al., 2001; Sakai et al., 2010), suggests novel treatment options. Prominent among these, the novel technology of reprogramming skin cells into pluripotent stem cells (iPSCs), already applied to EB (Bilousova et al., 2011; Itoh et al., 2011; Tolar et al., 2010) by a combination of specific transcription factors has the dual potential of generating patient-specific, highly proliferative cells for gene-correction strategies and of providing a tool for better understanding the biology of JEB-H. We hypothesized that such iPSCs can be derived from JEB-H individuals. Thus, in principle, an inexhaustible supply of patient-specific stem cells can be generated for local wound therapy and for systemic administration aimed at reaching both skin and internal mucosal membranes. To investigate this, we obtained skin biopsies from two individuals with JEB-H, who carried mutations in the LAMB3 gene: patient 1 P1; c.1365_1366del (p.Asn456ArgfsX7); c.2207C>A (p.Ser736X(Varki et al., 2006)) and patient 2 (P2; c.1903 C>T (p.Arg635X); c.1117C>T (p.Gln373X). Samples were obtained with written informed consent and following a protocol approved by the University of Minnesota Institutional Review Board and with adherence to the Helsinki Guidelines. All mutations create premature stop codons in the open reading frame, with expected nonsense-mediated decay of the mRNA or truncation of the protein product. The children experienced extensive areas of mucocutaneous lesions from birth, hoarseness and stridor, numerous infections, and progressive severe malnutrition. Examination of skin sections revealed the absence of laminin β3 chain at the DEJ. Electron microscopy examination showed infrequent and underdeveloped hemidesmosomes. Collectively these molecular, clinical, biochemical, and ultrastructural features were consistent with the diagnosis of JEB-H. To derive JEB-iPSCs, we transduced skin fibroblasts with the four transcription factors OCT4, SOX2, KLF4, and c-MYC, which are known to induce pluripotency in somatic cells(Tolar et al., 2010). Within three weeks of culture, the patient-specific JEB-iPSCs emerged as raised clusters of cells (Figure 1 a–c). To document the embryonic stem cell-like cellular state, we examined their mRNA and protein expression patterns. When compared with the parental fibroblasts, the JEB-iPSCs expressed the genes coding for nuclear, cytoplasmic, and cell surface proteins (e.g., TRA-1-60, TRA-1-81, stage-specific embryonic antigens 3 and 4, Lin28, Rex1, ABCG2 and DNMT3b, OCT4, and NANOG) in a pattern consistent with embryonic stem cell and iPSC phenotype (Figure 1 d–m). In support of the known activation of endogenous expression of stem cell genes by exogenous reprogramming factors, the maintenance of pluripotency became independent of the original exogenous reprogramming factors (data not shown). As expected in fully reprogrammed iPSCs, the epigenetic profiles showed that endogenous OCT4 and NANOG promoters were largely demethylated (Supplementary Figure 1). The JEB-iPSC lines were maintained for more than 20 passages, and they showed no evidence of genomic instability as evidenced by cytogenetic analysis (Supplementary Figure 2). To exclude the possibilities of cell contamination or the mosaicism observed in JEB(Pasmooij et al., 2007), we verified the authenticity of the JEB-iPSCs by genomic finger-typing with competitive polymerase chain reaction of a variable number of tandem repeat polymorphisms and by sequencing of LAMB3 gene mutations in the JEB-iPSCs (data not shown). To show that JEB-iPSCs are capable of differentiating into cells of endodermal, mesodermal, and ectodermal origin, we injected them into immune-deficient mice lacking T cells, B cells, and natural killer cells, and having a macrophage defect that makes them reliable recipients of human cells. In 6–8 weeks, cystic teratomas formed and cells derived from all three embryonic layers were seen (Figure 1 n). In aggregate, these data show that fully reprogrammed iPSCs can be derived from skin cells of JEB-H individuals. Figure 1 Expression profile of JEB-iPSCs JEB-iPSCs can provide means for drug screening and to model cellular interactions among various mucocutaneous cell types derived from the same individual. To our knowledge, this is a previously unreported use of iPSCs as a cellular tool to study the skin pathology in JEB. We showed first that skin-like structures formed in the process of in vivo JEB-iPSC differentiation (Supplementary Figure 3). In contrast to wild-type iPSCs, the skin-like structures arising from the JEB-iPSCs expressed no detectable laminin β3, but expressed collagen type VII, the DEJ protein deficient in distinct, dystrophic forms of EB (Figure 2 a–d). Next, to substantiate the proof-of-concept that skin cell cultures can be derived from JEB-iPSCs, we differentiated JEB-iPSCs into keratinocytes (Supplementary Figures 4 and 5). Lastly, to demonstrate that this operating procedure can serve as a platform for gene correction of these highly proliferative cells, we transduced the JEB-iPSCs with LAMB3 gene. After transduction, the JEB-iPSC-derived cells expressed and secreted laminin β3 protein (Figure 2 e). Figure 2 Skin cells derived from JEB-iPSCs In summary, we have shown that the LAMB3 defect does not preclude reprogramming into pluripotency, as has been observed in other genetic diseases(Raya et al., 2009). We have also shown that the JEB-iPSCs—in addition to establishing a reliable stem cell source for gene therapy interventions in JEB-H—can be used in the study of early human skin formation and compared to LM-332 in early development. With the ultimate clinical application of iPSC technology in mind, it is worth noting that strategies exist for genome-nonintegrating reprogramming, for depletion of tumor-inducing cells from differentiated iPSC cultures, and—as an JEB-H individual can develop anti-LM-332 antibody—for induction of immunological tolerance to disease-correcting transgenes(Vailly et al., 1998; Wu and Hochedlinger, 2011). Thus, gene-corrected JEB-iPSCs can inform medical advances in this severe and lethal blistering disease, as well as additional extracellular matrix disorders of the skin and other tissues(McGowan and Marinkovich, 2000).


Circulation-heart Failure | 2015

Derivation and High Engraftment of Patient-Specific Cardiomyocyte-Sheet Using Induced Pluripotent Stem Cells Generated From Adult Cardiac Fibroblast

Liying Zhang; Jing Guo; Pengyuan Zhang; Qiang Xiong; Steven C. Wu; Lily Xia; Samit S. Roy; Jakub Tolar; Timothy D. O’Connell; Michael Kyba; K. Liao; Jianyi Zhang

Background—Induced pluripotent stem cells (iPSCs) can be differentiated into potentially unlimited lineages of cell types for use in autologous cell therapy. However, the efficiency of the differentiation procedure and subsequent function of the iPSC-derived cells may be influenced by epigenetic factors that the iPSCs retain from their tissues of origin; thus, iPSC-derived cells may be more effective for treatment of myocardial injury if the iPSCs were engineered from cardiac-lineage cells, rather than dermal fibroblasts. Methods and Results—We show that human cardiac iPSCs (hciPSCs) can be generated from cardiac fibroblasts and subsequently differentiated into exceptionally pure (>92%) sheets of cardiomyocytes (CMs). The hciPSCs passed through all the normal stages of differentiation before assuming a CM identity. When using the fibrin gel–enhanced delivery of hciPSC-CM sheets at the site of injury in infarcted mouse hearts, the engraftment rate was 31.91%±5.75% at Day 28 post transplantation. The hciPSC-CM in the sheet also appeared to develop a more mature, structurally aligned phenotype 28 days after transplantation and was associated with significant improvements in cardiac function, vascularity, and reduction in apoptosis. Conclusions—These data strongly support the potential of hciPSC-CM sheet transplantation for the treatment of heart with acute myocardial infarction.


npj Regenerative Medicine | 2016

CRISPR/Cas9-based genetic correction for recessive dystrophic epidermolysis bullosa

Beau R. Webber; Mark J. Osborn; Amber N. McElroy; Kirk Twaroski; Cara-lin Lonetree; Anthony P. Defeo; Lily Xia; Cindy R. Eide; Christopher J. Lees; Ron T. McElmurry; Megan Riddle; Chong Jai Kim; Dharmeshkumar Patel; Bruce R. Blazar; Jakub Tolar

Recessive dystrophic epidermolysis bullosa (RDEB) is a severe disorder caused by mutations to the COL7A1 gene that deactivate production of a structural protein essential for skin integrity. Haematopoietic cell transplantation can ameliorate some of the symptoms; however, significant side effects from the allogeneic transplant procedure can occur and unresponsive areas of blistering persist. Therefore, we employed genome editing in patient-derived cells to create an autologous platform for multilineage engineering of therapeutic cell types. The clustered regularly interspaced palindromic repeats (CRISPR)/Cas9 system facilitated correction of an RDEB-causing COL7A1 mutation in primary fibroblasts that were then used to derive induced pluripotent stem cells (iPSCs). The resulting iPSCs were subsequently re-differentiated into keratinocytes, mesenchymal stem cells (MSCs) and haematopoietic progenitor cells using defined differentiation strategies. Gene-corrected keratinocytes exhibited characteristic epithelial morphology and expressed keratinocyte-specific genes and transcription factors. iPSC-derived MSCs exhibited a spindle morphology and expression of CD73, CD90 and CD105 with the ability to undergo adipogenic, chondrogenic and osteogenic differentiation in vitro in a manner indistinguishable from bone marrow-derived MSCs. Finally, we used a vascular induction strategy to generate potent definitive haematopoietic progenitors capable of multilineage differentiation in methylcellulose-based assays. In totality, we have shown that CRISPR/Cas9 is an adaptable gene-editing strategy that can be coupled with iPSC technology to produce multiple gene-corrected autologous cell types with therapeutic potential for RDEB.

Collaboration


Dive into the Lily Xia's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jakub Tolar

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Megan Riddle

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Scott Bell

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Troy C. Lund

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge