Network


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

Hotspot


Dive into the research topics where Cindy R. Eide is active.

Publication


Featured researches published by Cindy R. Eide.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Somatic mosaicism in Fanconi anemia: Evidence of genotypic reversion in lymphohematopoietic stem cells

John J. Gregory; John E. Wagner; Peter C. Verlander; Orna Levran; Sat Dev Batish; Cindy R. Eide; Amy Steffenhagen; Betsy Hirsch; Arleen D. Auerbach

Somatic mosaicism has been observed previously in the lymphocyte population of patients with Fanconi anemia (FA). To identify the cellular origin of the genotypic reversion, we examined each lymphohematopoietic and stromal cell lineage in an FA patient with a 2815–2816ins19 mutation in FANCA and known lymphocyte somatic mosaicism. DNA extracted from individually plucked peripheral blood T cell colonies and marrow colony-forming unit granulocyte–macrophage and burst-forming unit erythroid cells revealed absence of the maternal FANCA exon 29 mutation in 74.0%, 80.3%, and 86.2% of colonies, respectively. These data, together with the absence of the FANCA exon 29 mutation in Epstein–Barr virus-transformed B cells and its presence in fibroblasts, indicate that genotypic reversion, most likely because of back mutation, originated in a lymphohematopoietic stem cell and not solely in a lymphocyte population. Contrary to a predicted increase in marrow cellularity resulting from reversion in a hematopoietic stem cell, pancytopenia was progressive. Additional evaluations revealed a partial deletion of 11q in 3 of 20 bone marrow metaphase cells. By using interphase fluorescence in situ hybridization with an MLL gene probe mapped to band 11q23 to identify colony-forming unit granulocyte–macrophage and burst-forming unit erythroid cells with the 11q deletion, the abnormal clone was exclusive to colonies with the FANCA exon 29 mutation. Thus, we demonstrate the spontaneous genotypic reversion in a lymphohematopoietic stem cell. The subsequent development of a clonal cytogenetic abnormality in nonrevertant cells suggests that ex vivo correction of hematopoietic stem cells by gene transfer may not be sufficient for providing life-long stable hematopoiesis in patients with FA.


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.


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.


Stem Cells Translational Medicine | 2016

Rapid Induction of Cerebral Organoids From Human Induced Pluripotent Stem Cells Using a Chemically Defined Hydrogel and Defined Cell Culture Medium

Beth A. Lindborg; John H. Brekke; Amanda L. Vegoe; Connor Ulrich; Kerri T. Haider; Sandhya Subramaniam; Scott L. Venhuizen; Cindy R. Eide; Paul J. Orchard; Weili Chen; Qi Wang; Francisco Pelaez; Carolyn M. Scott; Efrosini Kokkoli; Susan A. Keirstead; James R. Dutton; Jakub Tolar; Timothy D. O’Brien

Tissue organoids are a promising technology that may accelerate development of the societal and NIH mandate for precision medicine. Here we describe a robust and simple method for generating cerebral organoids (cOrgs) from human pluripotent stem cells by using a chemically defined hydrogel material and chemically defined culture medium. By using no additional neural induction components, cOrgs appeared on the hydrogel surface within 10–14 days, and under static culture conditions, they attained sizes up to 3 mm in greatest dimension by day 28. Histologically, the organoids showed neural rosette and neural tube‐like structures and evidence of early corticogenesis. Immunostaining and quantitative reverse‐transcription polymerase chain reaction demonstrated protein and gene expression representative of forebrain, midbrain, and hindbrain development. Physiologic studies showed responses to glutamate and depolarization in many cells, consistent with neural behavior. The method of cerebral organoid generation described here facilitates access to this technology, enables scalable applications, and provides a potential pathway to translational applications where defined components are desirable.


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.


Leukemia | 2013

CXCR4 expression in CD34+ cells and unit predominance after double umbilical cord blood transplantation

Pablo Ramirez; John E. Wagner; Todd E. DeFor; Cindy R. Eide; Jeffrey S. Miller; Daniel J. Weisdorf; Claudio G. Brunstein

CXCR4 expression in CD34+ cells and unit predominance after double umbilical cord blood transplantation


Laboratory Investigation | 2017

Rapid generation of Col7a1 −/− mouse model of recessive dystrophic epidermolysis bullosa and partial rescue via immunosuppressive dermal mesenchymal stem cells

Beau R. Webber; Kyle T O'Connor; Ron T. McElmurry; Elise N Durgin; Cindy R. Eide; Christopher J. Lees; Megan Riddle; Wendy Mathews; Natasha Y. Frank; Mark A. Kluth; Christoph Ganss; Branden S. Moriarity; Markus H. Frank; Mark J. Osborn; Jakub Tolar

Recessive dystrophic epidermolysis bullosa (RDEB) is a debilitating and ultimately lethal blistering disease caused by mutations to the Col7a1 gene. Development of novel cell therapies for the treatment of RDEB would be fostered by having immunodeficient mouse models able to accept human cell grafts; however, immunodeficient models of many genodermatoses such as RDEB are lacking. To overcome this limitation, we combined the clustered regularly interspaced short palindromic repeats and associated nuclease (CRISPR/Cas9) system with microinjection into NOD/SCID IL2rγcnull (NSG) embryos to rapidly develop an immunodeficient Col7a1−/− mouse model of RDEB. Through dose optimization, we achieve F0 biallelic knockout efficiencies exceeding 80%, allowing us to quickly generate large numbers of RDEB NSG mice for experimental use. Using this strategy, we clearly demonstrate important strain-specific differences in RDEB pathology that could underlie discordant results observed between independent studies and establish the utility of this system in proof-of-concept human cellular transplantation experiments. Importantly, we uncover the ability of a recently identified skin resident immunomodulatory dermal mesenchymal stem cell marked by ABCB5 to reduce RDEB pathology and markedly extend the lifespan of RDEB NSG mice via reduced skin infiltration of inflammatory myeloid derivatives.


Advanced Materials | 2018

3D Printed Functional and Biological Materials on Moving Freeform Surfaces

Zhijie Zhu; Shuang Zhuang Guo; Tessa Hirdler; Cindy R. Eide; Xiaoxiao Fan; Jakub Tolar; Michael C. McAlpine

Conventional 3D printing technologies typically rely on open-loop, calibrate-then-print operation procedures. An alternative approach is adaptive 3D printing, which is a closed-loop method that combines real-time feedback control and direct ink writing of functional materials in order to fabricate devices on moving freeform surfaces. Here, it is demonstrated that the changes of states in the 3D printing workspace in terms of the geometries and motions of target surfaces can be perceived by an integrated robotic system aided by computer vision. A hybrid fabrication procedure combining 3D printing of electrical connects with automatic pick-and-placing of surface-mounted electronic components yields functional electronic devices on a free-moving human hand. Using this same approach, cell-laden hydrogels are also printed on live mice, creating a model for future studies of wound-healing diseases. This adaptive 3D printing method may lead to new forms of smart manufacturing technologies for directly printed wearable devices on the body and for advanced medical treatments.


Transplantation | 1987

The successful allotransplantation of neonatal rat islets across multiple combined major and minor histocompatibility barriers

Janet R. Serie; Orion D. Hegre; Cindy R. Eide; Anthony J. Weinhaus; Sue Marshall

Cultured neonatal rat islets were transplanted across six strain combinations into nonimmunosuppressed allogeneic recipients. Islets were isolated nonenzymatically by an in vitro method and were cultured at 37°C in 5% CO2 in air for 10 days prior to transplant. Transplants to nondiabetic recipients across four allogeneic barriers resulted in morphologically intact and wellgranulated islet tissue present at the graft site in 54 of 55 cases for periods lasting as long as 445 days (mean day of sacrifice was 163). In trials using diabetic recipients, ACIs receiving WF islets (n = 3) and outbred Holtzmans receiving Holtzman islets (n = 3) were reversed and did not return to the hyperglycemic state for experimental periods of up to 430 days.

Collaboration


Dive into the Cindy R. Eide's collaboration.

Top Co-Authors

Avatar

Jakub Tolar

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Bruce R. Blazar

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lily Xia

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Megan Riddle

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ron T. McElmurry

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge