ƽ

STEMdiff™ Hematopoietic Kit

For differentiation of human ES or iPS cells into hematopoietic progenitor cells

Need a high-quality cell source? Choose from our hiPSC healthy control lines, manufactured with mTeSR™ Plus.

STEMdiff™ Hematopoietic Kit

For differentiation of human ES or iPS cells into hematopoietic progenitor cells

Catalog #
(Select a product)
For differentiation of human ES or iPS cells into hematopoietic progenitor cells
Request Pricing Request Pricing

What's Included

  • STEMdiff™ Hematopoietic Basal Medium, 120 mL
  • STEMdiff™ Hematopoietic Supplement A (200X), 225 µL
  • STEMdiff™ Hematopoietic Supplement B (200X), 375 µL

What Our Scientist Says

We developed the STEMdiff™ Hematopoietic Kit so scientists like you can have a reliable source of hPSC-derived hematopoietic progenitor cells for their research.

Marta WalasekSenior Scientist
Marta Walasek, Senior Scientist

Overview

Generate functional hematopoietic progenitor cells from human embryonic stem (ES) and induced pluripotent stem (iPS) cells with STEMdiff™ Hematopoietic Kit. Using a simple and reproducible protocol in serum- and feeder-free conditions, you can produce hematopoietic progenitor cells expressing CD34, CD45, and CD43.

The robust, two-stage protocol initially induces cells toward the mesoderm before further differentiating them into hematopoietic progenitor cells. After 12 days, a population of hematopoietic cells containing 25 - 65% (average 43%) CD34+CD45+ progenitor cells can be harvested, including progenitor cells that have the capacity to form functional hematopoietic colonies in the colony-forming unit (CFU) assay.

STEMdiff™ Hematopoietic Kit has been optimized for differentiation of human pluripotent cells maintained in:
ձ𳧸™1 (Catalog #85850)
mTeSR™ Plus (Catalog #100-0276)
ձ𳧸™-8™ (Catalog #05990)
Subtype
Specialized Media
Cell Type
Hematopoietic Stem and Progenitor Cells, Pluripotent Stem Cells
Species
Human
Application
Cell Culture, Differentiation
Brand
STEMdiff
Area of Interest
Stem Cell Biology
Formulation Category
Serum-Free

Data Figures

Hematopoietic Cell Differentiation Protocol

Figure 1. Hematopoietic Differentiation Protocol

On Day -1, harvest and seed human ES/iPS cell colonies as small aggregates in ձ𳧸™1, mTeSR™ Plus, or TeSR™-E8. After one day, TeSR™ medium is replaced with Medium A (STEMdiff™ Hematopoietic Basal Medium containing Supplement A) to begin inducing the cells towards a mesoderm-like state (day 0). On day 2, a half medium change is performed with fresh Medium A. On day 3, the medium is changed to Medium B (STEMdiff™ Hematopoietic Basal Medium containing Supplement B) with half medium changes on days 5, 7 and 10, to promote further differentiation into hematopoietic cells. Typically, by day 12, large numbers of HPCs can be harvested from the culture supernatant.

Morphology of hPSC-Derived HPCs

Figure 2. Morphology of hPSC-Derived HPCs

Representative images of (A) hES (H1) cells and (B) hiPS (WLS-1C) cells on Day 12 of differentiation to HPCs using the STEMdiff™ Hematopoietic Kit. Differentiated cells exhibit typical HPC morphology as round cells that float freely in suspension.

Efficient and Robust Generation of CD34+CD45+/CD43+ HPCs
Efficient and Robust Generation of CD34+CD45+/CD43+ HPCs
Efficient and Robust Generation of CD34+CD45+/CD43+ HPCs

Figure 3. Efficient and Robust Generation of CD34+CD45+/CD43+ HPCs

hES and hiPS cells were cultured for 12 days in single wells of 12-well plates using the STEMdiff™ Hematopoietic Kit. At the end of the culture period, cells in suspension were harvested and analyzed by flow cytometry for expression of hematopoietic cell surface markers: CD34, CD45 and CD43. (A,B) Example flow cytometry plots for hematopoietic cell surface-marker analysis of cultures of hES (H1 and H9) and hiPS (STiPS-M001) cells. (C,D) Percentages and total numbers of CD34+CD45+ cells in cultures of hES (H1 and H9) or hiPS (WLS-1C, STiPS-F016, STiPS-M001 and STiPS-B004) cells are shown. Data shown as mean ± SEM; n ≥ 3.

hPSC-Derived HPCs Produce Colonies of Multiple Lineages
hPSC-Derived HPCs Produce Colonies of Multiple Lineages
hPSC-Derived HPCs Produce Colonies of Multiple Lineages

Figure 4. hPSC-Derived HPCs Produce Colonies of Multiple Lineages

Cells in suspension were harvested from the cultures on Day 12 of the hematopoietic differentiation protocol and assessed in colony-forming unit (CFU) assays using MethoCult™ H4435 Enriched (Catalog #04435) methylcellulose-based medium. (A) CFU frequencies in cultures of 6 different hPSC lines. (Data shown as mean ± SEM; n ≥ 3.) The CFU frequencies were variable between the different cell lines, with on average approximately 120 CFU per 10,000 hPSC-derived HPCs plated. (B) The progenitor cell types observed included granulocyte/macrophage (CFU-M, CFU-G and CFU-GM), erythroid (BFU-E and CFU-E) and occasional mixed (CFU-GEMM) colonies. Representative colony images are shown at 40X magnification.

Density plots showing CD34+ and CD45+ expression and percentage of cells co-expressing CD34+ and CD45+ and graphs showing total number of viable cells harvested.

Figure 5. Generation of Hematopoietic Progenitor Cells from hPSCs Maintained in mTeSR™ Plus

Human ES (H1, H9) and iPS (STiPS-M001, WLS-1C) cell lines maintained in ձ𳧸™1 (daily feeds) or mTeSR™ Plus (restricted feeds) were differentiated to hematopoietic progenitor cells using the STEMdiff™ Hematopoietic Kit. At the end of the differentiation period, cells were harvested from the supernatant and analyzed by flow cytometry for co-expression of CD34+ and CD45+ . (A) Representative density plots showing CD34+ and CD45+ expression, (B) percentage of cells co-expressing CD34+ and CD45+ , and (C) total number of viable cells harvested are shown. Data are expressed as the mean (± SEM); n=4.

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
Document Type
Product Name
Catalog #
05310
Lot #
All
Language
English
Document Type
Product Name
Catalog #
05310
Lot #
All
Language
English
Document Type
Product Name
Catalog #
05310
Lot #
All
Language
English
Document Type
Product Name
Catalog #
05310
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 (30)

A neuroimmune cerebral assembloid model to study the pathophysiology of familial Alzheimer’s disease A. Becerra-Calixto et al. Journal of Neuroinflammation 2025 Oct

Abstract

Alzheimer’s disease (AD) is the leading cause of dementia globally. The accumulation of amyloid and tau proteins, neuronal cell death and neuroinflammation are seen with AD progression, resulting in memory and cognitive impairment. Microglia are crucial for AD progression as they engage with neural cells and protein aggregates to regulate amyloid pathology and neuroinflammation. Recent studies indicate that microglia contribute to the propagation of amyloid beta (Aβ) via their immunomodulatory functions including Aβ phagocytosis and inflammatory cytokine production. Three-dimensional cell culture techniques provide the opportunity to study pathophysiological changes in AD in human-derived samples that are difficult to recapitulate in animal models (e.g., transgenic mice). However, these models often lack immune cells such as microglia, which play a critical role in AD pathophysiology. In this study, we developed a neuroimmune assembloid model by integrating cerebral organoids (COs) with induced microglia-like cells (iMGs) derived from human induced pluripotent stem cells from familial AD patient with PSEN2 mutation. After 120 days in culture, we found that iMGs were successfully integrated within the COs. Interestingly, our assembloids displayed histological, functional and transcriptional features of the pro-inflammatory environment seen in AD, including amyloid plaque-like and neurofibrillary tangle-like structures, reduced microglial phagocytic capability, and enhanced neuroinflammatory and apoptotic gene expression. In conclusion, our neuroimmune assembloid model effectively replicates the inflammatory phenotype and amyloid pathology seen in AD. The online version contains supplementary material available at 10.1186/s12974-025-03544-x.
Novel regulators of heparan sulfate proteoglycans modulate cellular uptake of α-synuclein fibrils B. Vanderperre et al. Communications Biology 2025 Oct

Abstract

Synucleinopathies are characterized by the accumulation and propagation of α-synuclein (α-syn) aggregates throughout the brain, leading to neuronal dysfunction and death. In this study, we used an unbiased FACS-based genome-wide CRISPR/Cas9 knockout screening to identify genes that regulate the entry and accumulation of α-syn preformed fibrils (PFFs) in cells. We identified key genes and pathways specifically implicated in α-syn PFFs intracellular accumulation, including heparan sulfate proteoglycans (HSPG) biosynthesis and Golgi trafficking. All confirmed hits affected heparan sulfate (HS), a post-translational modification known to act as a receptor for proteinaceous aggregates including α-syn and tau. Intriguingly, deletion of SLC39A9 and C3orf58 genes, encoding respectively a Golgi-localized exporter of Zn 2+ , and the Golgi-localized putative kinase DIPK2A, specifically impaired the uptake of α-syn PFFs, by preventing the binding of PFFs to the cell surface. Mass spectrometry-based analysis of HS chains in SLC39A9 -/- and C3orf58 -/- cells indicated major defects in HS homeostasis. Additionally, Golgi accumulation of NDST1, a prime HSPG biosynthetic enzyme, was detected in C3orf58 -/- cells. Interestingly, C3orf58 -/- human iPSC-derived microglia and dopaminergic neurons exhibited a strong reduction in their ability to internalize α-syn PFFs. Altogether, our data identifies new modulators of HSPGs that regulate α-syn PFFs cell surface binding and uptake. Subject terms: Cellular neuroscience, Glycobiology
Alzheimer's disease–associated PLCG2 variants alter microglial state and function in human induced pluripotent stem cell–derived microglia‐like cells L. M. Bedford et al. Alzheimer's & Dementia 2025 Oct

Abstract

Variants of phospholipase C gamma 2 (PLCG2), a key microglial immune signaling protein, are genetically linked to Alzheimer's disease (AD) risk. Understanding how PLCG2 variants alter microglial function is critical for identifying mechanisms that drive neurodegeneration or resiliency in AD. Induced pluripotent stem cell (iPSC) –derived microglia carrying the protective PLCG2 P522R or risk‐conferring PLCG2 M28L variants, or loss of PLCG2, were generated to ascertain the impact on microglial transcriptome and function. Protective PLCG2 P522R microglia showed significant transcriptomic similarity to isogenic controls. In contrast, risk‐conferring PLCG2 M28L microglia shared similarities with PLCG2 KO microglia, with functionally reduced TREM2 expression, blunted inflammatory responses, and increased proliferation and cell death. Uniquely, PLCG2 P522R microglia showed elevated cytokine secretion after lipopolysaccharide (LPS) stimulation and were protected from apoptosis. These findings demonstrate that PLCG2 variants drive distinct microglia transcriptomes that influence microglial functional responses that could contribute to AD risk and protection. Targeting PLCG2‐mediated signaling may represent a powerful therapeutic strategy to modulate neuroinflammation. The impact of Alzheimer's disease protective‐ and risk‐associated variants of phospholipase C gamma 2 (PLCG2) on the transcriptome and function of induced pluripotent stem cell (iPSC) –derived microglia was investigated. PLCG2 risk variant microglia exhibited a basal transcriptional profile similar to PLCG2‐deficient microglia but significantly different from isotype control and the transcriptionally similar PLCG2 protective variant microglia. PLCG2 risk variant and PLCG2‐deficient microglia show decreased levels of triggering receptor expressed on myeloid cells 2 (TREM2). The differential transcriptional pathways of protective and risk‐associated PLCG2 variant microglia functionally affect proliferation, apoptosis, and immune response. Protective PLCG2 microglia show resilience to apoptosis and increased cytokine/chemokine secretion upon exposure to lipopolysaccharide (LPS).
Need a high-quality cell source? Choose from our hiPSC healthy control lines, manufactured with mTeSR™ Plus.