Shi-Jiang Lu's research while affiliated with Ocata Therapeutics and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (79)


Human ESC-Derived MSCs Outperform Bone Marrow MSCs in the Treatment of an EAE Model of Multiple Sclerosis
  • Article
  • Full-text available

February 2021

·

83 Reads

·

24 Citations

Stem Cell Reports

·

·

Kumiko Ijichi

·

[...]

·

Ren-He Xu
Download
Share


Genome-wide DNA methylation drives human embryonic stem cell erythropoiesis by remodeling gene expression dynamics

November 2017

·

35 Reads

·

5 Citations

Epigenomics

Epigenomics

Aim: To investigate the role of DNA methylation during erythrocyte production by human embryonic stem cells (hESCs). Methods: We employed an erythroid differentiation model from hESCs, and then tracked the genome-wide DNA methylation maps and gene expression patterns through an Infinium HumanMethylation450K BeadChip and an Ilumina Human HT-12 v4 Expression Beadchip, respectively. Results: A negative correlation between DNA methylation and gene expression was substantially enriched during the later differentiation stage and was present in both the promoter and the gene body. Moreover, erythropoietic genes with differentially methylated CpG sites that were primarily enriched in nonisland regions were upregulated, and demethylation of their gene bodies was associated with the presence of enhancers and DNase I hypersensitive sites. Finally, the components of JAK-STAT-NF-κB signaling were DNA hypomethylated and upregulated, which targets the key genes for erythropoiesis. Conclusion: Erythroid lineage commitment by hESCs requires genome-wide DNA methylation modifications to remodel gene expression dynamics.


Retinal vascular injuries and intravitreal human embryonic stem cell-derived haemangioblasts

June 2017

·

10 Reads

·

1 Citation

Acta Ophthalmologica

Objective: To investigate whether intravitreally applied haemangioblasts (HB) derived from human embryonic stem cells (hESCs) are helpful for the repair of vascular damage caused in animals by an oxygen-induced retinopathy (OIR), by an induced diabetic retinopathy (DR) or by an induced retinal ischaemia with subsequent reperfusion. Methods: Human embryonic stem cell-derived HBs were transplanted intravitreally into C57BL/6J mice (OIR model), into male Wistar rats with an induced DR and into male Wistar rats undergoing induced retinal ischaemia with subsequent reperfusion. Control groups of animals received an intravitreal injection of endothelial cells (ECs) or phosphate-buffered saline (PBS). We examined the vasculature integrity in the mice with OIR, the blood-retina barrier in the rats with induced DR, and retinal thickness and retinal ganglion cell density in retina flat mounts of the rats with the retinal ischaemic-reperfusion retinopathy. Results: In the OIR model, the study group versus control groups showed a significantly (p < 0.001) smaller retinal avascular area [5.1 ± 2.7%;n = 18 animals versus 12.2 ± 2.8% (PBS group; n = 10 animals) and versus 11.8 ± 3.7% (EC group; n = 8 animals)] and less retinal neovascularization [6.3 ± 2.5%;n = 18 versus 15.2 ± 6.3% (n = 10; PBS group) and versus 15.8 ± 3.3% (n = 8; EC group)]. On retinal flat mounts, hESC-HBs were integrated into damaged retinal vessels and stained positive for PECAM (CD31) as EC marker. In the DR model, the study group versus the EC control group showed a significantly (p = 0.001) better blood-retina barrier function as measured at 2 days after the intravitreal injections [study group: 20.2 ± 12.8 μl/(g × hr); n = 6; versus EC control group: 52.9 ± 9.9 μl/(g × hr; n = 6)]. In the retinal ischaemia-reperfusion model, the groups did not differ significantly in retinal thickness and retinal ganglion cell density at 2, 5 and 7 days after baseline. Conclusion: By integrating into damaged retinal vessels and differentiating into ECs, intravitreally administered hESC-HBs may have partially repaired a retinal vascular injury caused by OIR model and DR.


Figure 1: In vitro Differentiation of Human Embryonic Stem Cells towards Retinal Neural Progenitors.: (a) Immunofluorescence staining shows co-expression of PAX6 and RX1 on day 13 eye field progenitors. (b) Quantification of PAX6 and RX1double positive eye field progenitors by flow cytometry analysis which shows >90% of them expressing both PAX6 and RX1 proteins. (c) Phase contrast image shows neural rosette structures of retinal neuronal progenitor cells (RNPC, far left) and immunofluorescence staining shows expression of PAX6 and CHX10 on RNPC at about day 30 after initial differentiation in vitro. Scale bar, 50 μm.
Figure 2: In vitro Differentiation of Retinal Neural Progenitors towards Photoreceptor-like Progenitors.: (a) Immunofluorescence staining shows the expression of transcription factors NRL, NR2E3 and CRX in PhRPs at 90 –100 days after in vitro differentiation. CHX10 and neuN genes are negative in PhRPs at this stage; the upper right corner of the CHX10 image shows positive expression of CHX10 in RNPCs at day 30 (positive control); the upper right corner of the neuN image shows positive expression of neuN in mouse central nerve cells (positive control). 2nd antibody only also shows negative staining. (b) Quatification of intracellular staining of NRL, NR2E3 and CRX as determined by Flow Cytometry analyses. Scale bar, 50 μm.
Figure 3: In vitro Generation of Mature Photoreceptor-like Cells from Human ESC/iPS-Derived PhRPs.: Expression of rod photoreceptor markers, rhodopsin, recoverin and PDE6α in hESC (a) and iPSC-derived (b) photoreceptor-like cells two week after in vitro maturation. Scale bar, 50 μm.
Figure 4: Transplanted Human ESC-PhRPs and iPSC-PhRPs Survive in the Subretinal Space of rd1 Mice.: Scanning laser ophthalmoscopy (SLO) was performed in vivo three weeks post transplantation to assess the extent of surviving donor cells; GFP positive cells are observed in autofluorescence (AF) mode as white dots or clusters (black areas represent areas of retina which were not seeded with transplanted cells, due to incomplete detachment of the retina around the optic nerve head). Representative near-infrared (NIR) and AF fundus images of rd1 mice show a homogeneous presence of GFP+ cells in the two treatment groups: ESC-PhRPs (a) and iPSC-PhRPs (b). Histological assessment 3 weeks post transplantation revealed ESC-PhRP (c-c’) and iPSC-PhRP (d-d’) derived cell layers (green) between the retinal pigment epithelium (RPE) and inner nuclear layer (INL) of the rd1 retina, replacing the absent outer nuclear layer (ONL) in the adult rd1 mice; (e-e’) GFP+ cells were stained with human nuclear antigen (HNA) which co-localized with GFP; indicating that the GFP signal observed in vivo in treated animals was indeed an indicator of transplanted human PhRPs. Scale bar, 25 μm.
Figure 5: Transplanted Human ESC-PhRPs and iPSC-PhRPs Express Mature Photoreceptor Markers in vivo.: Immunofluorescence staining 3 weeks post transplantation shows expression of mature photoreceptor markers in transplanted human PhRPs (green). In all images cells are located in the subretinal space and oriented so that the host INL is located at the top of the image and the RPE at the bottom. The pan-photoreceptor marker recoverin was observed within the reconstructed layer of cells in animals treated with both ESC-PhRPs (a) and iPSC-PhRPs (b,c). The rod specific enzyme phosphodiesterase β6 (PDE6b), which is necessary in phototransduction and is absent in rd1 mice due to mutation was reinstated in the retina and located in the outer processes of transplanted ESC-PhRPs (d-d’) and iPSC-PhRPs (e-e’). The rod specific protein rhodopsin, which is normally located in outer segment membrane disk was also observed in outer segments of ESC-PhRPs (f) and iPSC-PhRPs (g). Cone arrestin was observed in GFP+ cells, indicating that a subset of human cells matured to produce cone photoreceptors. Scale bar, 20 μm.

+2

Function of human pluripotent stem cell-derived photoreceptor progenitors in blind mice

July 2016

·

633 Reads

·

135 Citations

Scientific Reports

Photoreceptor degeneration due to retinitis pigmentosa (RP) is a primary cause of inherited retinal blindness. Photoreceptor cell-replacement may hold the potential for repair in a completely degenerate retina by reinstating light sensitive cells to form connections that relay information to downstream retinal layers. This study assessed the therapeutic potential of photoreceptor progenitors derived from human embryonic and induced pluripotent stem cells (ESCs and iPSCs) using a protocol that is suitable for future clinical trials. ESCs and iPSCs were cultured in four specific stages under defined conditions, resulting in generation of a near-homogeneous population of photoreceptor-like progenitors. Following transplantation into mice with end-stage retinal degeneration, these cells differentiated into photoreceptors and formed a cell layer connected with host retinal neurons. Visual function was partially restored in treated animals, as evidenced by two visual behavioral tests. Furthermore, the magnitude of functional improvement was positively correlated with the number of engrafted cells. Similar efficacy was observed using either ESCs or iPSCs as source material. These data validate the potential of human pluripotent stem cells for photoreceptor replacement therapies aimed at photoreceptor regeneration in retinal disease.



Generation of Hemangioblasts from Human Pluripotent Stem Cells

August 2015

·

32 Reads

·

1 Citation

Hemangioblasts are progenitors with the capacity to differentiate into hematopoietic, endothelial, smooth muscle, and mesenchymal stromal cells and represent an excellent candidate of cell therapy for a variety of human diseases. To realize their clinical potential, first, an efficient and controlled differentiation toward hemangioblasts in a scalable manner, probably in a bioreactor setting, from an unlimited source is required. These cells also need to be generated under animal components and cell-free conditions, or additional regulatory challenges, such as xeno-transplant, will have to be addressed. The ability of human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSC) to divide indefinitely without losing pluripotency may allow them to serve as an inexhaustible source for the large-scale production of therapeutic cells. In this chapter, we describe a robust system that can efficiently generate large numbers of hemangioblasts from multiple hESCs and iPSC lines under well-defined conditions, which is an important step for future clinical applications, with the potential of developing a GMP-compatible scalable system.


Transdifferentiation of Human Hair Follicle Mesenchymal Stem Cells into Red Blood Cells by OCT4

February 2015

·

500 Reads

·

22 Citations

Shortage of red blood cells (RBCs, erythrocytes) can have potentially life-threatening consequences for rare or unusual blood type patients with massive blood loss resulting from various conditions. Erythrocytes have been derived from human pluripotent stem cells (PSCs), but the risk of potential tumorigenicity cannot be ignored, and a majority of these cells produced from PSCs express embryonic ε- and fetal γ-globins with little or no adult β-globin and remain nucleated. Here we report a method to generate erythrocytes from human hair follicle mesenchymal stem cells (hHFMSCs) by enforcing OCT4 gene expression and cytokine stimulation. Cells generated from hHFMSCs expressed mainly the adult β-globin chain with minimum level of the fetal γ-globin chain. Furthermore, these cells also underwent multiple maturation events and formed enucleated erythrocytes with a biconcave disc shape. Gene expression analyses showed that OCT4 regulated the expression of genes associated with both pluripotency and erythroid development during hHFMSC transdifferentiation toward erythroid cells. These findings show that mature erythrocytes can be generated from adult somatic cells, which may serve as an alternative source of RBCs for potential autologous transfusion.



Blood Components from Pluripotent Stem Cells

December 2014

·

44 Reads

Limitations in the supply of transfusable blood components can have potentially life-threatening consequences for patients with massive blood loss, or who suffer from diseases that cause severe anemia. The ability of human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) to divide indefinitely without losing pluripotency and to theoretically differentiate into any cell type in the body makes them highly attractive cell sources for large-scale regenerative medicine purposes. Many researchers have already been able to differentiate hESCs and/or iPSCs into specific mature blood cell types, including erythroid, myeloid, macrophage, megakaryocyte/platelet and lymphoid cell lineages. For example, hESC-derived red blood cells and platelets are functional in tasks such as oxygen delivery and blood clotting, respectively, and may be able to serve as substitutes for their donor-derived counterparts in emergencies. hESC-derived dendritic cells are functional in antigen-presentation and may be used as off-the-shelf vaccine therapies to stimulate antigen-specific immune responses against cancer cells. The ability to create banks of hESC lines with matched or reduced incompatibility could potentially reduce or eliminate the need for immunosuppressive drugs and/or immunomodulatory protocols altogether, for example, (O) RhD− lines for generation of universal red blood cells. Inasmuch as human iPSCs could potentially be produced from a patient’s own cells, all types of blood cells derived from such iPSC lines would be histocompatible with the patient. For transfusion-dependent patients with unusual or rare blood types, particularly those who are alloimmunized, cells generated in this manner could be an invaluable source of transfusable cells. In this chapter, we highlight the biological functions of mature cells of the blood, clinical conditions requiring the transfusion or stimulation of these cells, and the potential for hESC/iPSC derivatives to serve as functional replacements. However, in vitro differentiation systems used to generate these cells will need further optimization before hESC/iPSC-derived blood components can be used clinically.


Citations (36)


... However, in these reports, MSCs were all produced in 2D cultures and required several purification and manipulation steps, thus hampering mass production. Traditional monolayer cultures allow the external control of targeted hiPSC differentiation to produce more uniform cell populations [12,[41][42][43]. However, these cultures lack the 3D cell assembly properties that could define endogenous biological systems. ...

Reference:

Mesenchymal stem/stromal cells from human pluripotent stem cell-derived brain organoid enhance the ex vivo expansion and maintenance of hematopoietic stem/progenitor cells
Human ESC-Derived MSCs Outperform Bone Marrow MSCs in the Treatment of an EAE Model of Multiple Sclerosis

Stem Cell Reports

... The second wave of erythropoiesis arises following switching from the yolk sac to the fetal liver in the second gestational month in humans, in conjunction with generation of smaller "definitive" erythroid cells in circulation and the switch of hemoglobin from embryonic (Iturri et al., 2021;S. J. Lu & Lanza, 2019;S. J. Lu, Park, Feng, & Lanza, 2009). ...

Cell Therapy for Blood Substitutes
  • Citing Chapter
  • January 2019

... Although the ESCs are commonly derived from ICM of blastocysts, single blastomere biopsied from 2-to 8-cell stages of pre-implantation embryos has also shown to be able to form ESC colonies (18)(19)(20). Furthermore, embryonal germ cells isolated from genital ridges of post-implantation embryos are also capable of forming ESC colonies (21,22). ...

Human Embryonic Stem Cell Lines Derived from Single Blastomeres
  • Citing Article
  • January 2007

Obstetrical and Gynecological Survey

... We found that approximately 69% of DMCs associated with CpG islands were located in promoter regions ( Table 2). Liu et al. (Liu et al., 2017) reported that the proportions of hypermethylated CpG sites located in CpG islands, CpG shores and other locations were 25.49∼34.23%, 21.57∼40.75%, ...

Genome-wide DNA methylation drives human embryonic stem cell erythropoiesis by remodeling gene expression dynamics
  • Citing Article
  • November 2017

Epigenomics

Epigenomics

... However, the subretinal injection procedure is considered to be a complicated and risky operation, which is prone to retinal detachment, cataracts, vitreous hemorrhage, recurrence of submacular hemorrhage, and postoperative development of CNV and other incurable injuries [20]. In contrast, the intravitreal injection procedure is established and widely used in clinical treatment of retinal diseases [4,[21][22][23][24]. Additionally, the vitreous cavity is a relevantly immune-privileged site with a low risk of immune rejection. ...

Retinal vascular injuries and intravitreal human embryonic stem cell-derived haemangioblasts
  • Citing Article
  • June 2017

Acta Ophthalmologica

... In contrast, chemical induction avoids the use of genetic factors; therefore, future studies should explore whether hCiPSCs could bypass the potential safety issues associated with overexpression of genetic factors. Second, considering the complex, multilayered structure of the retina and the involvement of its diverse cell layers in disease progression, exclusive reliance on the replacement of dysfunctional RPE cells may not achieve optimal therapeutic effects 30, 31 . Therefore, future studies should explore alternative hCiPSC-based approaches such as co-transplantation of RPE cells with retinal progenitor cells derived from hCiPSCs, as well as the transplantation of more complex and structured hCiPSC-derived multilayered retinal cells or organoids 32 . ...

Function of human pluripotent stem cell-derived photoreceptor progenitors in blind mice

Scientific Reports

... Human HPSC engraftment in the spleen and lymph node at the 8th week [9,139,143] Mouse and human iPSCs HSCs iPSCs co injected with OP9 stromal cells into mice to form teratomas. HSCs were taken from the bone marrow From mouse iPSCs blood cells engraftment at the 4th and 12th weeks From Human iPSCs erythrocytes and CD3+ T cells engraftment at 8 weeks [9,139,144] Human iPSCs Hemangioblasts Culture on the MEF feeder layer Not assayed [9,139,145] Human iPSCs Hemangioblasts 3D culture using Matrigel-coated microcarrier. Not assayed [9,146,147] GFi1B: Growth factor independent 1B transcriptional repressor, a protein-coding gene. ...

Generation of Hemangioblasts from Human Pluripotent Stem Cells
  • Citing Chapter
  • August 2015

... The second wave of erythropoiesis arises following switching from the yolk sac to the fetal liver in the second gestational month in humans, in conjunction with generation of smaller "definitive" erythroid cells in circulation and the switch of hemoglobin from embryonic (Iturri et al., 2021;S. J. Lu & Lanza, 2019;S. J. Lu, Park, Feng, & Lanza, 2009). ...

Red Blood Cells
  • Citing Chapter
  • December 2009

... In our earlier research, we discovered that human hair follicle mesenchymal stem cells (hHFMSCs) derived from the bulge and papilla of hair follicles can be induced into iPSCs by the introduction of four pluripotent factors (Oct4, Sox2, c-Myc, and Klf4) [3] . Additionally, we found that introducing only OCT4 can reprogram hHFMSCs into intermediate cells (hHFMSCs OCT4 ), and these reprogrammed cells can be used to generate mature erythrocytes directly by stimulating them with speci c cytokines [4] . Furthermore, we observed an intriguing phenomenon in which pre-stimulation with low concentrations of FLT-3 and SCF cytokines resulted in the generation of suspended cell subsets capable of in vitro passage and expansion, as well as delaying differentiation. ...

Transdifferentiation of Human Hair Follicle Mesenchymal Stem Cells into Red Blood Cells by OCT4

... With rapid advances in biological and gene editing technology, HLA-universal platelets, which refer to blood components with reduced or even no HLA antigen expression and that can be supplied continuously, have been successfully produced in vitro. 70,71 It is an appealing prospect for transfusion medicine that will not only alleviate the resource deficit but also prevent HLA alloimmunization and refractoriness. Several starting cells including CD34 + hematopoietic progenitor cells, 72 pluripotent stem cells (PSCs), induced human pluripotent stem cells (iPSCs), 70,73 and embryonic stem cells (ESCs) 74 have been extensively studied as an ex vivo source of platelet manufacture. ...

Scalable Generation of Universal Platelets from Human Induced Pluripotent Stem Cells

Stem Cell Reports