Vitronectin XF™

Defined, xeno-free matrix that supports the growth and differentiation of human pluripotent stem cells under serum-free, feeder-free conditions

Vitronectin XF™

Defined, xeno-free matrix that supports the growth and differentiation of human pluripotent stem cells under serum-free, feeder-free conditions

From: 87 USD
Catalog #
(Select a product)
Defined, xeno-free matrix that supports the growth and differentiation of human pluripotent stem cells under serum-free, feeder-free conditions
Add to Wish List

Product Advantages


  • Defined, recombinant human protein

  • Consistent, reproducible human ES and iPS cell cultures

  • Convenient, room temperature plate coating

  • Enzyme-free passaging

  • Defined system when used with TeSR™-E8™, mTeSR™1, or TeSR™2

Overview

Support the growth and differentiation of human pluripotent stem cells by using Vitronectin XF™, a defined and xeno-free cell culture matrix.

Developed and manufactured by Nucleus Biologics, Vitronectin XF™ is an effective alternative to Corning® Matrigel®. Use Vitronectin XF™ with mTeSR™1, mTeSR™ Plus, TeSR™-E8™, or TeSR™-AOF medium to provide a defined culture system for the maintenance of human embryonic stem (ES) cells and human induced pluripotent stem (iPS) cells under feeder-free conditions. This system allows complete control over the culture environment, resulting in more consistent cell populations and reproducible results in downstream applications.

Note: CellAdhere™ Dilution Buffer and non-tissue culture-treated cultureware (e.g. Catalog #100-0096/27147) are required for use with Vitronectin XF™ and are available for purchase separately.
Cell Type
Endoderm, PSC-Derived, Pluripotent Stem Cells
Species
Human

Data Figures

Figure 1. Morphology of Human ES and iPS Cells Cultured on Vitronectin XF™ Cell Culture Matrix in TeSR™-E8™

Undifferentiated human ES (H9) and iPS (WLS-1C) cell cultures exhibit normal morphology when cultured on Vitronectin XF™. Colonies are round, tightly packed and multilayered, with a high nucleus-to-cytoplasm ratio. Cells were transferred directly from Matrigel® hESC-Qualified Matrix without an adaptation step. Note: Colonies grown in TeSR™-E8™ have a more condensed and round morphology when grown on Vitronectin XF™ matrix, compared to colonies grown on Matrigel® hESC-Qualified Matrix, which are more diffuse and irregularly shaped.

Protocols and Documentation

Find supporting information and directions for use in the Product Information Sheet or explore additional protocols below.

Document Type
Product Name
Catalog #
Lot #
Language
Product Name
Vitronectin XF™
Catalog #
100-0763, 07180
Lot #
All
Language
English
Document Type
Safety Data Sheet
Product Name
Vitronectin XF™
Catalog #
100-0763, 07180
Lot #
All
Language
English

Applications

This product is designed for use in the following research area(s) as part of the highlighted workflow stage(s). Explore these workflows to learn more about the other products we offer to support each research area.

Resources and Publications

Publications (4)

Non-integrating episomal plasmid-based reprogramming of human amniotic fluid stem cells into induced pluripotent stem cells in chemically defined conditions. Slamecka J et al. Cell cycle (Georgetown, Tex.) 2016 JAN

Abstract

Amniotic fluid stem cells (AFSC) represent an attractive potential cell source for fetal and pediatric cell-based therapies. However, upgrading them to pluripotency confers refractoriness toward senescence, higher proliferation rate and unlimited differentiation potential. AFSC were observed to rapidly and efficiently reacquire pluripotency which together with their easy recovery makes them an attractive cell source for reprogramming. The reprogramming process as well as the resulting iPSC epigenome could potentially benefit from the unspecialized nature of AFSC. iPSC derived from AFSC also have potential in disease modeling, such as Down syndrome or $\$-thalassemia. Previous experiments involving AFSC reprogramming have largely relied on integrative vector transgene delivery and undefined serum-containing, feeder-dependent culture. Here, we describe non-integrative oriP/EBNA-1 episomal plasmid-based reprogramming of AFSC into iPSC and culture in fully chemically defined xeno-free conditions represented by vitronectin coating and E8 medium, a system that we found uniquely suited for this purpose. The derived AF-iPSC lines uniformly expressed a set of pluripotency markers Oct3/4, Nanog, Sox2, SSEA-1, SSEA-4, TRA-1-60, TRA-1-81 in a pattern typical for human primed PSC. Additionally, the cells formed teratomas, and were deemed pluripotent by PluriTest, a global expression microarray-based in-silico pluripotency assay. However, we found that the PluriTest scores were borderline, indicating a unique pluripotent signature in the defined condition. In the light of potential future clinical translation of iPSC technology, non-integrating reprogramming and chemically defined culture are more acceptable.
Functionalizing Ascl1 with Novel Intracellular Protein Delivery Technology for Promoting Neuronal Differentiation of Human Induced Pluripotent Stem Cells Robinson M et al. Stem Cell Reviews and Reports 2016 AUG

Abstract

Pluripotent stem cells can become any cell type found in the body. Accordingly, one of the major challenges when working with pluripotent stem cells is producing a highly homogenous population of differentiated cells, which can then be used for downstream applications such as cell therapies or drug screening. The transcription factor Ascl1 plays a key role in neural development and previous work has shown that Ascl1 overexpression using viral vectors can reprogram fibroblasts directly into neurons. Here we report on how a recombinant version of the Ascl1 protein functionalized with intracellular protein delivery technology (Ascl1-IPTD) can be used to rapidly differentiate human induced pluripotent stem cells (hiPSCs) into neurons. We first evaluated a range of Ascl1-IPTD concentrations to determine the most effective amount for generating neurons from hiPSCs cultured in serum free media. Next, we looked at the frequency of Ascl1-IPTD supplementation in the media on differentiation and found that one time supplementation is sufficient enough to trigger the neural differentiation process. Ascl1-IPTD was efficiently taken up by the hiPSCs and enabled rapid differentiation into TUJ1-positive and NeuN-positive populations with neuronal morphology after 8 days. After 12 days of culture, hiPSC-derived neurons produced by Ascl1-IPTD treatment exhibited greater neurite length and higher numbers of branch points compared to neurons derived using a standard neural progenitor differentiation protocol. This work validates Ascl1-IPTD as a powerful tool for engineering neural tissue from pluripotent stem cells.
Chemically defined conditions for human iPSC derivation and culture. Chen G et al. Nature methods 2011 MAY

Abstract

We re-examine the individual components for human embryonic stem cell (ESC) and induced pluripotent stem cell (iPSC) culture and formulate a cell culture system in which all protein reagents for liquid media, attachment surfaces and splitting are chemically defined. A major improvement is the lack of a serum albumin component, as variations in either animal- or human-sourced albumin batches have previously plagued human ESC and iPSC culture with inconsistencies. Using this new medium (E8) and vitronectin-coated surfaces, we demonstrate improved derivation efficiencies of vector-free human iPSCs with an episomal approach. This simplified E8 medium should facilitate both the research use and clinical applications of human ESCs and iPSCs and their derivatives, and should be applicable to other reprogramming methods.