Reprogramming method for efficient inducing of T cells into multipotent stem cells

A technology for pluripotent stem cells and reprogramming, applied in animal cells, vertebrate cells, cell culture active agents, etc., can solve the problem of iPSC preparation technology failing to achieve large-scale preparation, and achieve the effect of high immune cell differentiation efficiency

Active Publication Date: 2018-10-12
安徽中盛溯源生物科技有限公司
View PDF3 Cites 5 Cited by
  • Summary
  • Abstract
  • Description
  • Claims
  • Application Information

AI Technical Summary

Problems solved by technology

[0013] These findings indicate that the use of iPSC technology to manipulate T cells may be very useful for the development of more effectiv

Method used

the structure of the environmentally friendly knitted fabric provided by the present invention; figure 2 Flow chart of the yarn wrapping machine for environmentally friendly knitted fabrics and storage devices; image 3 Is the parameter map of the yarn covering machine
View more

Image

Smart Image Click on the blue labels to locate them in the text.
Viewing Examples
Smart Image
  • Reprogramming method for efficient inducing of T cells into multipotent stem cells
  • Reprogramming method for efficient inducing of T cells into multipotent stem cells
  • Reprogramming method for efficient inducing of T cells into multipotent stem cells

Examples

Experimental program
Comparison scheme
Effect test

Example Embodiment

[0070] Example 1: Design and construction of episomal vector expression system

[0071] Such as figure 2 As shown, in this example, three episomal vectors were constructed, all of which were amplified by direct polymerase chain reaction (PCR) from some or all open reading frames (ORF) (using coding regions) from pluripotency determining factor genes. The first and last 20-22 bases are used as primers), and the above ORF is inserted into the commercially available mammalian expression vector pCEP4 or the related backbone containing OriP / EBNA1 to generate an episomal vector. All three episomal vectors contain at least An internal ribosome entry size (IRES), the first vector is pEP4-E-O2S-E-N2K( figure 2 A), which in turn contains the first promoter, POU5F1, IRES2, SOX2, the second promoter, NANOG, IRES2 and KLF4; the second vector is pEP4-E-O2S-E-T2K( figure 2 B), which in turn contains the third promoter, POU5F1, IRES2, SOX2, the fourth promoter, SV40LT, IRES2 and KLF4; the thir...

Example Embodiment

[0072] Example 2: Primary isolation, culture and identification of T cell monocytes

[0073] 1. Primary isolation and culture of T cell monocytes

[0074] Collect a 6ml blood sample, transfer it to a lymphocyte separation tube, centrifuge, take the mononuclear cell layer, wash twice with DPBS, sample and count, and use flow cytometry to detect the expression of the surface molecules CD3+ and CD4+CD8+. Detect the positive rate and average fluorescence intensity indicators separately (the experimental results are as image 3 A), take 6×10 according to the counting result 6 Cells were seeded in 3 wells in a 6-well plate coated with activator, 2ml / well of T cell serum-free medium was added, and placed at 37°C, 5% CO 2 The culture in the incubator constitutes the expansion culture system of the present invention, and each well is supplemented with 1 to 2 ml of fresh T cell serum-free medium on the first day of expansion.

[0075] The serum-free medium for T cells in this embodiment includ...

Example Embodiment

[0080] Example 3: Episomal vector induces reprogramming

[0081] 1. Method

[0082] a. Take 0.5~4×10 T cells activated and cultured for 2 days in Example 2 6 , Using the pEP4-E-O2S-E-N2K, pEP4-E-O2S-E-T2K and pCEP4-LM-2L episomal vectors constructed in Example 1 to electrotransfect target cells, and then inoculate them in hiPSC induction medium and Matrigel Or cultured in a six-well plate coated with vitronectin or other cell matrix, the transfection content of each plasmid DNA is pEP4-E-O2S-E-N2K: pEP4-E-O2S-E-T2K: pCEP4-LM -2L=1:1:1.

[0083] b. After 48 hours, replace the fresh hiPSC induction medium with half the amount, continue to culture for 10 days, and change the medium the next day, that is, reprogram on the feeder-free system.

[0084] The specific components of the pluripotent stem cell induction medium are:

[0085] One or more of the following small molecules were added per liter of the serum-free expansion medium for T cells in Example 2: CHIR99021 1 μmol, A-83-01 0.5 μ...

the structure of the environmentally friendly knitted fabric provided by the present invention; figure 2 Flow chart of the yarn wrapping machine for environmentally friendly knitted fabrics and storage devices; image 3 Is the parameter map of the yarn covering machine
Login to view more

PUM

No PUM Login to view more

Abstract

The invention discloses a reprogramming method for efficient inducing of T cells into multipotent stem cells. The method comprises the following steps: S1, extracting monocyte from a blood sample, andadding the monocyte to an amplification culture system containing an activating agent for implementing selective T cell activating culture; S2, introducing an episomal vector which contains at leastone potential determinant into the T cells which are obtained in the step S1; S3, cultivating the T cells which are obtained in the step S2 and contain the episomal vector via a multipotent stem cellinducing medium, and performing inducing in a system free from a feeding layer, so that preprogrammed intermediate cells are obtained; and S4, after complete inducing, replacing the multipotent stem cell inducing medium in the step S3 with a multipotent stem cell medium for maintaining culture, so that cells that the expression of the potential determinant disappears and expression of endogenous multipotent genes, namely POU5F1, NANOG, TRA-1-60 and TRA-1-81, is activated are obtained, wherein the cells are induced multipotent stem cells. The method provided by the invention has the beneficialeffect that the T-cell derived induced multipotent stem cells can be simply and conveniently prepared in a large scale.

Description

technical field [0001] The invention belongs to the field of cells, and in particular relates to a reprogramming method for efficiently inducing T cells into pluripotent stem cells. Background technique [0002] In general, stem cells are undifferentiated cells capable of giving rise to a succession of mature, functional cells. Embryonic stem cells (ESCs), for example, are derived from embryos and are pluripotent, thus having the ability to develop into any organ or tissue type in the adult. Hematopoietic stem cells can give rise to any of the different types of terminally differentiated blood cells. Induced pluripotent stem cells (induced pluripotent stem cells), usually referred to as iPS cells or iPSCs, are a type of pluripotent stem cells artificially derived from non-pluripotent cells (usually adult cells). iPSCs were first generated from mouse cells in 2006 (Takahashi et al., 2006) and from human cells in 2007 (Takahashi et al., 2007; Yu et al., 2007). The study of ...

Claims

the structure of the environmentally friendly knitted fabric provided by the present invention; figure 2 Flow chart of the yarn wrapping machine for environmentally friendly knitted fabrics and storage devices; image 3 Is the parameter map of the yarn covering machine
Login to view more

Application Information

Patent Timeline
no application Login to view more
IPC IPC(8): C12N15/85C12N5/10
CPCC12N5/0696C12N15/85C12N2506/115C12N2510/00C12N2501/608C12N2501/604C12N2501/606C12N2501/605C12N2501/602C12N2501/60
Inventor 俞君英周桃陈涛涛董成友张颖
Owner 安徽中盛溯源生物科技有限公司
Who we serve
  • R&D Engineer
  • R&D Manager
  • IP Professional
Why Eureka
  • Industry Leading Data Capabilities
  • Powerful AI technology
  • Patent DNA Extraction
Social media
Try Eureka
PatSnap group products