AU2021106553A4 - Modification method for MFS Patient-specific Induced Pluripotent Stem Cells - Google Patents

Modification method for MFS Patient-specific Induced Pluripotent Stem Cells Download PDF

Info

Publication number
AU2021106553A4
AU2021106553A4 AU2021106553A AU2021106553A AU2021106553A4 AU 2021106553 A4 AU2021106553 A4 AU 2021106553A4 AU 2021106553 A AU2021106553 A AU 2021106553A AU 2021106553 A AU2021106553 A AU 2021106553A AU 2021106553 A4 AU2021106553 A4 AU 2021106553A4
Authority
AU
Australia
Prior art keywords
mfs
cells
patient
nucleofection
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
AU2021106553A
Inventor
Minjie Hu
Peifeng Jin
Yongyu Wang
Jiwen YANG
Huan ZHANG
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Wenzhou Medical University
Original Assignee
Wenzhou Medical University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Wenzhou Medical University filed Critical Wenzhou Medical University
Priority to AU2021106553A priority Critical patent/AU2021106553A4/en
Application granted granted Critical
Publication of AU2021106553A4 publication Critical patent/AU2021106553A4/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0696Artificially induced pluripotent stem cells, e.g. iPS
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • C12N2015/8527Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic for producing animal models, e.g. for tests or diseases
    • C12N2015/8536Animal models for genetic diseases
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/28Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from vascular endothelial cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/108Plasmid DNA episomal vectors

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Cell Biology (AREA)
  • Plant Pathology (AREA)
  • Molecular Biology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Developmental Biology & Embryology (AREA)
  • Transplantation (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The invention discloses a selection or modification method for MFS patient-specific induced pluripotent stem cell (iPSC), which belongs to the technical field of stem cells reprogramming and disease modelling; the method includes the following steps: episomal plasmids containing transcription factors are introduced into the primary vascular cells to be expressed by nucleofection.

Description

Modification method for MFS Patient-specific Induced Pluripotent Stem Cells
TECHNICAL FIELD
The invention relates to the field of iPSC reprogramming, in particular to a method for
generation of MFS patient-specific iPSC.
BACKGROUND
Marfan syndrome (MFS) is an autosomal dominant connective tissue disease with an
incidence of one in 3000 to 5000. In 90% of cases, it is caused by an alteration of Fibrillin
1 (FBN1) gene. FBNJ is a gene with 65 exons located on chromosome 15q-21.1. FBNJ is
a matrix glycoprotein and the main component of elastic fibers. Its changes can interfere
with the TGF-j signalling pathway. In addition to changes in the FBNJ gene, there may
also be changes in the gene encoding the TGF- receptor (TGF3R). Some individuals with
TGFRI or TGFPRII gene alterations have clinical features consistent with MFS, which
further proves the role of activating the TGF- signalling pathway in the pathogenesis of
MFS. Obtaining patient-specific iPSCs for MFS may provide a new in vitro human-derived
cell model for elucidating the molecular mechanism of MFS. So far, the research on the
molecular mechanism of MFS disease is not completely clear. At present, the research of
MFS is mainly carried out at the animal level. Due to species differences, animal models
and humans are very different in various physiological systems, and many research results
cannot be directly applied to its clinical application. Therefore, how to establish a human
derived iPSC and its differentiated vascular cell model is very important for the research
of cardiovascular diseases such as MFS.
The emergence of human induced pluripotent stem cells (hiPSCs) has provided a
revolutionary technology for translational medicine and regenerative medicine, as well as new methods and a large number of cell sources for the study of human genetic disease mechanisms. Initially, in 2006, Shinya Yamanaka at Kyoto University first reported the generation of mouse induced pluripotent stem cells in Cell. They cloned the four transcription factor genes Oct3/4, Sox2, c-Myc, and Klf4 into viral vectors, and then introduced them into mouse fibroblasts, with finding that generated iPS cells are similar to embryonic stem cells in terms of morphology, gene and protein expression, epigenetic modification status, cell multiplication ability, embryoid body and teratoma formation ability, and differentiation potential. In November 2007, the Thompson laboratory and the
Yamanaka laboratory almost simultaneously reported that the application of
reprogramming technology can also induce human skin fibroblasts to become iPSCs almost
identical to embryonic stem cells. Based on the generation and application of iPS cells, it
provides a new way to construct cells models, and the generation of MFS patient-specific
induced pluripotent stem cell models will provide useful cell models for studying the
disease.
SUMMARY
The purpose of the present invention is to provide a method for efficiently generating MFS
patient-specific induced pluripotent stem cells, which provides an inexhaustible cell source
for various type of cell differentiation for disease modelling, drug screening and other
application.
In order to achieve the objectives above, the present invention provides the following
solutions:
The present invention provides MFS patient-specific induced pluripotent stem cells which
includes the following steps:
The isolated vascular tissue from MFS patients is cultured in vitro to obtain primary
vascular cells;
The episomal plasmids containing the transcription factor are introduced into the primary
vascular cells for expression by a nucleofection method to obtain specific iPSC cell lines.
Preferably, the primary vascular cells are obtained according to the following steps:
Cut the obtained aortic tissue of the MFS patient into small pieces of about 1 mm3 , and
attach them to the culture flask, add DMEM medium containing 10% fetal bovine serum
and 1% penicillin/streptomycin (P/S), one side of attached the tissue blocks upward for a
period of culture, turn the culture flask over and continue culturing until a large number of
cells grew out and further expand to obtain primary vascular cells.
Preferably, the vascular cells include fibroblasts and vascular smooth muscle cells.
Preferably, obtaining the specific iPSC cell strain includes the following steps:
Step 1: When the vascular cells are cultured to have density of 8 0 - 9 0 %, they are digested
into single cells, centrifuged, and the cell pellet is taken;
Step 2: After mixing the episomal plasmids containing different genes with the
nucleofection solution, resuspending the cell pellet, and then performing nucleofection;
Step 3: After nucleofection, adding culture medium to resuspend the cells, and then
inoculating the cell suspension in DMEM medium containing 10% fetal bovine serum and
1% P/S. Changing the medium every day until the culture cell density is 90%. The cells
are digested, and cultured with the mixed solution-1. After one day of culture, changing to
the mixed solution-2. When specific iPSC clones are observed, changing to the culture
solution for iPSC clones' growth;
Step 4: After the iPSC clones are picked up, they are expanded and cultured with the
medium to obtain different iPSC cell lines.
Preferably, the mixed solution-i is a mixture of (hiPSC/hESC culture medium+10 ng/ml
HumanbFGF+0.25 mMNaB) and (DMEM+10% FBS+1% P/S medium) inavolume ratio
of 1:1;
The mixed solution-2 includes: hiPSC/hESC culture medium+10 ng/ml Human
bFGF+0.25 mM NaB;
The culture medium is: hiPSC/hESC culture medium.
Preferably, the episomal plasmids containing different genes include OCT4, KLF4, SOX2,
Lin28, 1-myc and shP53.
The invention also provides an iPSC model for MFS patients constructed according to the
reprogramming method.
The present invention also provides the application of the MFS patient iPSC model
constructed according to the reprogramming method in the following (a) or (b):
(a) Application in clinical drugs screening for treatment and/or prevention of MFS disease;
(b) Application in studying the cellular and molecular mechanisms of MFS-related
diseases.
The present invention discloses the following technical effects:
(1) The Marfan syndrome (MFS)-specific pluripotent stem cell (iPSC) disease model
established by the present invention has a patient-specific genetic background, which can
better reflect the patient's pathological state and the development process of the disease.
(2) At present, the establishment of iPSC mainly uses lentivirus (which will be integrated
into the genome), sendai virus (the preparation method is more difficult), etc. This study uses the episomal plasmids nucleofection method to directly transfer transcription factors into cells. This method is simple to operate and saves time. At the same time, genes will not be integrated into the genome, creating good conditions for MFS disease cells model.
BRIEF DESCRIPTION OF THE FIGURES
In order to explain the embodiments of the present invention or the technical solutions in
the prior art more clearly, the following will briefly introduce the figures needed in the
embodiments. Obviously, the figures in the following description are only some
embodiments of the present invention. For those of ordinary skill in the art, without creative
work, other figures can be obtained based on these figures.
Figure 1 shows the primary cultured aortic vascular cells of MFS patients; A: the
morphology of the primary cells that migrated and proliferated in the aorta tissue of MFS
patients; B: the morphology of the cells cultured after the passage of primary cells;
Figure 2 shows the reprogramming of primary aortic vascular cells from MFS patients into
iPSC; A: the cell morphology on Day 6 after nucleofection; B: small clones appeared on
Day 13 after nucleofection; C: On Day16 after nucleofection, small clones gradually grew
up; D: established passage 1 (P1) iPSC line in the culture plate;
Figure 3 shows the characterization of MFS patient-specific iPSCs; A: alkaline
phosphatase staining; B: karyotype analysis of MFS patient-specific iPSC; C:
immunofluorescence staining of pluripotency genes; D: PT-PCR products of pluripotency
genes in agarose gel; E: RT-qPCR quantified the expression of pluripotency gene; OCT4,
NANOG, SOX2, TRA-1-60, TRA-1-81 are pluripotency genes; Primary cell: primary
vascular cells from MFS patient aorta, H1ESC: HI human embryonic stem cell; F:
formation of embryonic body (EB); G: The staining of a-SMA, a marker of EB-mediated differentiation of mesodermal cell; H: The staining of a-Fetoprotein, a marker of EB mediated differentiation of endoderm cell; I: The staining of Nestin, a marker of EB mediated differentiation of ectoderm cell.
DESCRIPTION OF THE INVENTION
Various exemplary embodiments of the present invention will now be described in detail.
This part should not be considered as a limitation to the present invention, but as more
detailed information of certain aspects, characteristics, and embodiments of the present
invention.
It should be noted that the terms described in the present invention are only used to describe
specific embodiments and are not to limit the present invention. In addition, for the
numerical range in this invention, it should be understood that each intermediate value
between the maximum and the minimum is also specifically disclosed. Each smaller range
between any stated value or intermediate value within the stated range and any other stated
value or intermediate value within the stated range is also included in the present invention.
The upper and lower limits of these smaller ranges can be independently included or
excluded from the range.
Unless otherwise specified, all technical and scientific terms used herein have the same
meaning as commonly understood by those skilled in the art in the field of the present
invention. Although the present invention only describes preferred methods and materials,
any methods and materials similar or equivalent to those described herein can also be used
in the practice or testing of the present invention. All documents mentioned in this
specification are incorporated by reference to disclose and describe methods and/or materials related to the documents. In the event of conflict with any incorporated document, the content of this manual shall prevail.
Without departing from the scope or spirit of the present invention, various improvements
and changes can be made to the specific embodiments of the present specification, which
is apparent to those skilled in the art. Other embodiments derived from the description of
the present invention will be apparent to the skilled person. The specification and examples
of this application are only exemplary.
As used herein, "including", "composing", "having", "containing", etc., are all open terms,
which means including but not limited to.
Embodiment 1 A method for generation of MFS patient-specific pluripotent stem cells
includes the following steps:
1) Obtaining of primary vascular cells from patient aortic tissues
The tissue was obtained from a 28-year-old man from the First Affiliated Hospital of
Wenzhou Medical University. This patient was diagnosed as an MFS with hyaline
degeneration and mucous degeneration of the fibrous tissue of the "aortic valve". The
discarded aortic tissue of this patient was collected and rinsed with DPBS; then transferred
into dishes with DMEM basal medium; the tissue was cut into pieces with size of about 1
mm 3, which was attached to the bottom of the culture flask; the flask was placed up-and
down with culture medium (DMEM+10% FBS + 1% penicillin/streptomycin (P/S) in a
37C, 5% CO2 incubator for 2-3 hours; then the culture flask was turned over and the tissue
blocks could culture in the medium, and the medium changed every 2 days. When the
primary cells' density in the culture flask reached about 90%, which is referred to as PO primary cells (the morphology is shown in Figure 1A), and these cells were subcultured to further expansion, some of them were frozen for preservation.
Among them, the above-mentioned DPBS is a phosphate buffered saline, and DMEM is a
well-known medium containing various amino acids and glucose in the field. FBS is fetal
bovine serum, and the percentage in DMEM+10% FBS+1% penicillin/streptomycin (P/S)
medium is calculated by volume ratio.
2) Establishment of MFS patient-specific iPSC lines
After the primary cells' density has reached about 90% (the morphology is shown in Figure
IB), the cells were digested into single cells with 0.05% TE. Centrifugation was carried
out and the supernatant was aspirated. The four plasmids (containing the genes OCT4,
KLF4, SOX2, Lin28, 1-myc and shP53, purchased from Addgene, #27077, #27078,
#27080, #37624) and nucleofection solution (Lonza, #V4XP-3024) were mixed; the cell
pellet was resuspended, and then nucleofectionion was performed using LONZA 4D
Nucleofector with program FF-120.
After nucleofection, the culture solution was added to resuspend the cells; the cell
suspension was inoculated in medium containing DMEM+10% FBS+1% P/S; the medium
was changed every other day; on the 6th day,(Figure 2A),the cells' density is about 90 %.
On the 6th day, the digested cells were inoculated on a 35 mm culture dish pre-plated with
Matrigel, and the prepared culture medium for reprogramming (Nuwacell-ncEpic
hiPSC/hESC medium+10 ng/ml Human bFGF+0.25 mM NaB) mixed with culture medium
for primary cells (medium containing DMEM+10% FBS+1% P/S ) on the basis of 1:1 and
cultured for one day; then change the medium with the prepared medium for
reprogramming every other day. When iPSC clones emerged (Figure 2B), the medium should change to Nuwacell hiPSC/hESC medium (Nuwacell, #PR01001) daily. After the clones grow up (Figure 2C), they are picked up and expanded to establish specific iPSC cell lines.
Among them, Matrigel is from BD, and Nuwacell-ncEpic hiPSC/hESC culture medium is
from Nuwacell.
Episomal vector plasmids:
pCXLE-hOCT3/4-shp53 (Addgene, #27077)
pCXLE-hSK (Addgene, #27078)
pCXLE-hUL (Addgene, #27080)
pCXWB-EBNA1 (Addgene, #37624)
The nucleofection solution is from P3 Primary Cell 4D-Nucleofector X Kit L (Lonza,
#V4XP-3024).
The morphology of MFS patient-specific iPSCs during the reprogramming process is
shown in Figure 2. The results show that some cells initially underwent morphological
changes when they were cultured for 5 days after nucleofection, and some small iPSC
clones could be seen on day 13 after nucleofection. The clones will grow bigger after that.
The established MFS patient-specific iPSC lines will expand and stock (Figure 2D). The
iPSCs were characterized by small cells, high ratio of nucleus to cytoplasm, prominent
nuclei, and similar to ESC-like cells.
3) Characterization of MFS patient-specific iPSC lines
To characterize the pluripotency of established iPSC, the activity of alkaline phosphatase,
immunofluorescence staining and RT-PCR detection of pluripotency genes, embryoid
body (EB)-mediated differentiation of three germ layers, karyotype analysis were performed. The results showed that the iPSC clones showed very similar in cellular and molecular biology characteristics to that of hESCs. The detail procedures of these assays are as following:
The alkaline phosphatase staining (APS) assay identifies the pluripotency of iPSCs:
Follow the instructions of the BCIP/NBTAlkaline PhosphataseColor Development Kit for
APS.
The detailed process is as follows: aspirate the medium and wash the cells with PBS; fix
cells with 4% paraformaldehyde at room temperature for 20 minutes; then add the freshly
prepared APS working solution to stain cells for about 10 minutes. Stop the reaction,
aspirate the staining working solution, and wash twice with PBS. The positive APS staining
cells will show purple.
Immunofluorescence staining of pluripotency genes:
The results of immunofluorescence staining showed that pluripotency markers, OCT4,
NANOG, SOX2, expressed in the nucleus while TRA-1-60 and TRA-1-81 expressed on
the cell surface.
The process is as follows: the iPSC was cultured on a coverslip in 12-well plate, then fixed
cells with 4% paraformaldehyde for 20 minutes, then permeabilized and blocked the cells
in PBS containing 0. 5 % Triton X-100 and 5% normal goat serum for 1 hour at RT. The
cells incubate the following antibodies overnight at 4°C, rabbit anti-OCT4; rabbit anti
SOX2; rabbit anti-NANOG; mouse anti-TRA-1-60; mouse anti-TRA-1-81; mouse anti-a
SMA; rabbit anti-Nestin; rabbit anti-a-Fetoprotein. After wash with PBS, the cells stain
with secondary antibodies for 1 hour at room temperature, AF594 Goat Anti-Rabbit IgG;
AF488 Goat Anti-Mouse IgG; AF488 Goat Anti-Rabbit IgG. The cells were washed with ddH 20 and then mounted with a mounting medium containing 4',6-diamidino-2 phenylindole (DAPI), and then analyses using a Nikon microscope (Ts2-FL).
RT-PCR detection of pluripotency genes:
H1ESCs were used as a positive control, and primary vascular cells were used as a negative
control. The results showed that the MFS patient-specific iPSCs and the H1ESC were
clearly expressed the pluripotency markers, while the primary cells did not.
The process is as follows: cells were lysated with Trizol and total RNA was isolated;
PrimerScript RT Reagent Kit (Takara, RR037A) was used to reverse transcribe RNA into
cDNA; PCR was performed as standard procedure by adding specific primers and Taq
DNA polymerase. The amplified PCR products were detected by agarose gel
electrophoresis. The PCR product size of OCT4, SOX2 and NANOG gene is 138 bp, 397
bp and 811 bp.
RT-qPCR detection of pluripotency genes:
H1ESC was used as a positive control, and primary vascular cells were used as a negative
control. The results showed that the MFS patient-specific iPSCs and the H1ESC were
highly expressed the pluripotency markers while the primary vascular cells did not.
The specific process is as follows: Total RNA was isolated by Trizol and cDNA was
synthesized by PrimerScript RT Reagent Kit. qPCR was conducted in StepOne Plus Real
Time PCR System (Applied Biosystems) using iTaq Universal SYBR Green Supermix
(Bio-Rad) and specific primers for hESC pluripotency genes.
Table 1 PCR primers
Gene Forward/Reverse primer (5'-3') RT-PCR primers OCT4 (138bp) CATTCAAACTGAGGTAAGGG/ TAGCGTAAAAGGAGCAACATAG NANOG (811bp) TAGCAATGGTGTGACGCAGA/ CCTCGCTGATTAGGCTCCAA
SOX2 (397bp) ATGCACCGCTACGACG/ CTTTTGCACCCCTCCCATTT GAPDH (133bp) TAGCAATGGTGTGACGCAGA/ CCTCGCTGATTAGGCTCCAA qPCR primers OCT4 (60bp) AGTGCCCGAAACCCACACTG/ ACCACACTCGGACCACATCCT NANOG (134bp) TGAACCTCAGCTACAAACAG/ TGGTGGTAGGAAGAGTAAAG SOX2 (154bp) GCCGAGTGGAAACTTTTGTCG/GGCAGCGTGTACTTATCCTTCT GAPDH (80bp) TGTGGGCATCAATGGATTTGG/ ACACCATGTATTCCGGGTCAAT
Embryoid body (EB)-mediated differentiation of three germ layers:
The specific process is as follows: We differentiated MFS patient-specific iPSC cell lines
into embryoid bodies (EB) in low-attachment petri dishes. Embryoid bodies (EB) are
spherical structure with high morphological similarity to the early embryonic
developmental stages of mammals and contain different types of cells of the endoderm,
mesoderm and ectoderm. As shown in the figure, the markers of three germ layers -a
Fetoprotein (endoderm), a-SMA (mesoderm), Nestin (ectoderm) were all positive,
indicating that the MFS patient-specific iPSCs could differentiate into three germ layers.
The differentiation of EB:
The iPSCs with a density of about 80%-90% were treated with collagenase IV for 20-30
minutes and cultured in EB medium in ultra-low attachment dish for 6 days. The culture
was transferred in DMEM containing 20% FBS on a gelatin-coated 6-well plate for 6-12
days. The medium was changed every other day. The differentiated cells were fixed and
stained for markers of the three germ layers. The preparation of EB medium is shown in
Table 2.
Table 2 Preparation of EB medium
Component Amount Final Concentration DMEM/F12 77.8 mL -
KnockOut Serum 20ml 20% 100xMEM-Non-Essential amino acids iml 10 mM 1OOxL-Glutamine (DMEM/F12 lacks 1mi 20 mM glutamine) 55 mM 2-Mercaptoethanol or 0.1 M 182 jiL of 55 mM 0.1 mM (1000x) stock (0.1 mL of 0.1 M)
Karyotyping:
Karyotype analysis showed that the MFS patient-specific iPSCs had a normal karyotype.
Karyotyping was performed on G-band metaphase chromosomes with a resolution of 400
bands per haploid genome. Briefly, proliferating iPSCs were blocked by 50 ng/ml of
colcemid for 2 h, digested with trypsin-EDTA into single cell, then treated with hypotonic
KCl solution for 20-40 min at 37 °C. Glass slides were prepared with three steps of fixation
in methanol/glacial acetic acid (3:1). QFQ-banding at 400 bands resolution according to
the International System for Human Cytogenetic Nomenclature (ISCN2016) was analyzed.
The iPSCs were confirmed to display a normal karyotype (46, XY). The results of alkaline
phosphatase staining are shown in Figure 3A; the results of karyotype analysis of
chromosomes are shown in Figure 3B; the results of immunofluorescence staining are
shown in Figure 3C, and the RT-PCR results of pluripotency genes are shown in Figure
3D. The expression of pluripotency genes by RT-qPCR as shown in Figure 3E; the
identification of embryoid body (EB)-mediated differentiation of the three germ layers is
shown in Figure 3 (F, G, H, I).
4) MFS patient-specific iPSCs contain two mutations in FBN1 gene
The pathogenesis of Marfan syndrome (MFS) is not completely known, it appears to be
due to heterozygous mutation in the fibrillin-1 gene (FBN1; 134797) on chromosome q21. So far, 3077 loci of FBN1 have been genetically altered in the general database. We found the patient-specific iPSCs have two missense mutations, c.3442 C>G (p.P1148A) and c.1415 G>A (p.C472Y), in the FBN1 gene through whole-exome sequencing, which may be related to the pathogenesis of this disease.
The above-mentioned embodiments only describe the preferred mode of the present
invention, and do not limit the scope of the present invention. Without departing from the
design spirit of the present invention, variations and improvements to the technical
solutions of the present invention made by those of ordinary skill in the field should fall
within the protection scope determined by the claims of the present invention.

Claims (1)

  1. THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS
    1. A modification method for MFS patient-specific iPSC characterized in that it comprises
    the following steps:
    obtain a sample of isolated vascular tissue from MFS patients to obtain primary vascular
    cells;
    The episomal plasmids containing the transcription factors are introduced into the primary
    vascular cells by nucleofection.
    2. The method according to claim 1, wherein the obtaining of the primary vascular cells
    comprises the following steps:
    Cut the MFS patient aortic tissue into small pieces of about 1mm3 blocks, and attach them
    to the bottom of culture flask, place the flask up-and-down, then add DMEM medium
    containing 10% FBS and 1% P/S. Several hours later, flip the flask and let tissue merge in
    culture medium.
    3. The method according to claim 1, wherein the vascular cells include fibroblasts and
    vascular smooth muscle cells.
    4. The method according to claim 1, wherein the selecting of the specific iPSC lines
    comprises the following steps:
    Step 1: When the vascular cells are cultured to a cell density of 80-90%, they are digested
    into single cells, centrifuged, and the cell pellet is collected;
    Step 2: After mixing the episomal plasmids containing different genes with the
    nucleofection solution, resuspending the cell pellet, and then performing nucleofection;
    6. The method to claim 4, wherein the episomal plasmids containing transcription factors
    includes plasmids containing genes OCT4, KLF4, SOX2, Lin28, 1-myc and shP53.
    FIGURES 23 Aug 2021
    Figure 1 2021106553
    Figure 2
    Figure 3
AU2021106553A 2021-08-23 2021-08-23 Modification method for MFS Patient-specific Induced Pluripotent Stem Cells Active AU2021106553A4 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2021106553A AU2021106553A4 (en) 2021-08-23 2021-08-23 Modification method for MFS Patient-specific Induced Pluripotent Stem Cells

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
AU2021106553A AU2021106553A4 (en) 2021-08-23 2021-08-23 Modification method for MFS Patient-specific Induced Pluripotent Stem Cells

Publications (1)

Publication Number Publication Date
AU2021106553A4 true AU2021106553A4 (en) 2021-11-11

Family

ID=78488541

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2021106553A Active AU2021106553A4 (en) 2021-08-23 2021-08-23 Modification method for MFS Patient-specific Induced Pluripotent Stem Cells

Country Status (1)

Country Link
AU (1) AU2021106553A4 (en)

Similar Documents

Publication Publication Date Title
US20210324341A1 (en) Use of rna for reprogramming somatic cells
JP5497294B2 (en) Method for obtaining a xeno-free hBS cell line
AU2007271898B2 (en) A novel population of multipotent cardiac precursor cells derived from human blastocysts derived stem cells
US20040120932A1 (en) In vitro-derived adult pluripotent stem cells and uses therefor
CN102725399B (en) Materials and methods for generating pluripotent stem cells
US20130059378A1 (en) Human epidermis-derived mesenchymal stem cell-like pluripotent cells and preparation method thereof
CN112204149A (en) Reprogramming carrier
Bosch et al. Discovering the potential of dental pulp stem cells for corneal endothelial cell production: a proof of concept
WO2011016485A1 (en) METHOD FOR INDUCING DIFFERENTIATION OF iPS CELLS INTO HEPATIC PARENCHYMAL CELLS
AU2021106553A4 (en) Modification method for MFS Patient-specific Induced Pluripotent Stem Cells
Eminli et al. Clinically compatible advances in blood-derived endothelial progenitor cell isolation and reprogramming for translational applications
CA3199728A1 (en) Methods and cellular structures
CN109852587B (en) Preparation method of human Kekan syndrome specific adult stem cells
JP7198488B2 (en) Method for producing spheroid expressing pluripotent stem cell marker and method for expressing pluripotent stem cell marker
CN113430229B (en) Construction method of human-derived BAV-TAA disease pluripotent stem cell model
EP3875581A1 (en) Automated method for preparing keratinocytes
Mura et al. Generation of the human induced pluripotent stem cell (hiPSC) line PSMi007-A from a Long QT Syndrome type 1 patient carrier of two common variants in the NOS1AP gene
CN101517069A (en) A novel population of multipotent cardiac precursor cells derived from human blastocysts derived stem cells
KR100662706B1 (en) Culture method for human embryonic stem cells by using culture medium collected after amniotic fluid cells culture
KR102180733B1 (en) A Composition for Isolating Stem Cells Comprising Skim Milk as an Active Ingredient
WO2022268212A1 (en) Culture method for differentiating stem cells into cardiomyocytes, and culture medium
CN117242170A (en) Reprogramming somatic cells on microcarriers
Bekhite et al. Human Induced Pluripotent Stem Cell as a Disease Modeling and Drug Development Platform-A Cardiac Perspective. Cells 2021, 10, 3483
Baudyšová Isolated Organs and Explanted Tissue
KR100586462B1 (en) Culture method for human embryonic stem cells by using amniotic fluid cells

Legal Events

Date Code Title Description
FGI Letters patent sealed or granted (innovation patent)