Miniaturized 3D bone marrow tissue model to assess response to Thrombopoietin-receptor agonists in patients

  1. Christian A Di Buduo
  2. Pierre-Alexandre Laurent
  3. Carlo Zaninetti
  4. Larissa Lordier
  5. Paolo M Soprano
  6. Aikaterini Ntai
  7. Serena Barozzi
  8. Alberto La Spada
  9. Ida Biunno
  10. Hana Raslova
  11. James B Bussel
  12. David L Kaplan
  13. Carlo L Balduini
  14. Alessandro Pecci
  15. Alessandra Balduini  Is a corresponding author
  1. Department of Molecular Medicine, University of Pavia, Italy
  2. Department of Internal Medicine, I.R.C.C.S. San Matteo Foundation and the University of Pavia, Italy
  3. UMR 1170, Institut National de la Santé et de la Recherche Médicale, Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy Cancer Campus, Equipe Labellisée Ligue Nationale Contre le Cancer, France
  4. Integrated Systems Engineering, Italy
  5. Isenet Biobanking, Italy
  6. Institute for Genetic and Biomedical Research-CNR, Italy
  7. Department of Pediatrics, Weill Cornell Medicine, United States
  8. Department of Biomedical Engineering, Tufts University, United States
9 figures, 8 tables and 1 additional file

Figures

Figure 1 with 2 supplements
Design of the bone marrow mimicking device.

(A) To mimic the vascularized bone marrow tissue structure ex vivo a double-flow chamber device was designed in two parts. The core contains two separates flow channels dedicated to the perfusion having inlet and outlet ports for connection to a perfusion system. (B,C) The dimension of the polydimethylsiloxane (PDMS) mold cover top and (D,E) the core device is expressed in millimeters. Alternative models of the device are shown in Figure 1—figure supplement 1. The 3D-printed negative mold of the chamber is shown in Figure 1—figure supplement 2.

Figure 1—figure supplement 1
Various models of the silk bone marrow device.

The devices can be produced from one to four perfusion channels and molded with PDMS (scale bars = 10 mm).

Figure 1—figure supplement 2
3D printing of the negative mold.

(A) Drawing of negative molds for the core device (upper panel) and the cover tap (lower panel) are disposed in isometric view, right view, top view, and transversal section view, respectively. Dimensions are in millimeters. (B) The production is operated by an FDM 3D printer using PLA and (C) needles are finally mounted to connect the future inlet and outlet for perfusion of the chamber.

Silk sponge bone marrow perfusion system.

(A–C) A peristaltic pump drives perfusion of the cell culture medium from a reservoir to the device equipped with a silk fibroin sponge prepared directly inside the chamber by dispensing an aqueous silk solution mixed with salt particles (scale bar B = 1.5 cm; scale bar C = 2 mm). After leaching out the salt, the resulting porous silk sponge can be sterilized. (D,E) Confocal microscopy reconstruction of the silk sponge showed the presence of an interconnected alveolar network (scale bar D = 200 µm; scale bar E = 150 µm). (F) The analysis of pore diameters measured on the top and bottom of the scaffold demonstrated no significant differences throughout the scaffold. Results are presented as mean ± SD (n = 150 pore/condition, p=NS). (G) Confocal microscopy analysis of CFSE+ cells cultured within the silk scaffold (red = CFSE; gray = silk; scale bar = 50 µm). The full data set is provided in Figure 2—source data 1.

Figure 2—source data 1

Analysis of the pore diameter of the silk scaffolds.

https://cdn.elifesciences.org/articles/58775/elife-58775-fig2-data1-v1.xlsx
Figure 3 with 1 supplement
Modeling physiological and pathological megakaryopoiesis.

(A) Megakaryocytes were differentiated from healthy controls and patients affected by MYH9-RD and ANKRD26-RT patients and cultured into the bone marrow device in presence of 10 ng/mL TPO. (B) Output of CD41+CD42b+ megakaryocyte at the end of differentiation relative to healthy controls (n = 12 Healthy Controls, n = 12 MYH9-RD; n = 12 ANKRD26-RT) (C) Percentage of proplatelet formation relative to healthy controls (n = 12 Healthy Controls, n = 12 MYH9-RD; n = 12 ANKRD26-RT; *p<0.01). (D) The number of proplatelet bifurcation per single megakaryocytes in healthy controls and patients (n = 12 Healthy Controls, n = 12 MYH9-RD; n = 12 ANKRD26-RT; *p<0.01). (E) Representative immunofluorescence staining of proplatelet structure (red=β1-tubulin; blue = nuclei; scale bar = 20 µm). All results are presented as mean ± SD. Data from the treatment of healthy controls in the presence of TPO and TPO +EPAG are shown in Figure 3—figure supplement 1. The full data set is provided in Figure 3—source data 1.

Figure 3—source data 1

Analysis of megakaryocyte differentiation and proplatelet formation in healthy controls and patients.

https://cdn.elifesciences.org/articles/58775/elife-58775-fig3-data1-v1.xlsx
Figure 3—figure supplement 1
Eltrombopag is effective in promoting thrombopoiesis from healthy controls.

(A) Output of CD41+CD42b+ healthy control megakaryocytes at the end of differentiation cultured in the presence of TPO +EPAG, with respect to TPO alone (n = 6, *p<0.01). (B) The percentage of proplatelet forming-megakaryocytes was calculated as the number of cells displaying long filamentous pseudopods with respect to the total number of round megakaryocytes per analyzed field (n = 6; *p<0.01). All results are presented as mean ± SD. The full data set is provided in Figure 3—source data 1.

Figure 3—figure supplement 1—source data 1

Analysis of megakaryocyte output and proplatelet formation in healthy controls in the presence of EPAG.

https://cdn.elifesciences.org/articles/58775/elife-58775-fig3-figsupp1-data1-v1.xlsx
Eltrombopag promotes megakaryocyte differentiation ex vivo.

(A) Megakaryocytes were differentiated from peripheral blood progenitors of patients affected by MYH9-RD or ANKRD26-RT and cultured in the silk bone marrow tissue device in the presence of 10 ng/mL TPO supplemented or not with 500 ng/mL Eltrombopag (EPAG) and analyzed. The figure of the microscope was adapted from Servier Medical Art licensed under a Creative Commons Attribution 3.0 Unported License (https://smart.servier.com). (B) Representative immunofluorescence staining of CD61 (red = CD61; blue = nuclei; scale bar = 25 µm) and (C) analysis of ploidy levels at the end of the culture (TPO: n = 3 MYH9-RD; n = 3 ANKRD26-RT; TPO +EPAG: n = 3 MYH9-RD; n = 3 ANKRD26-RT; p=NS). (D) Representative flow cytometry analysis of CD41+CD42b+ megakaryocytes at the end of the culture and (E) statistical analysis of mean fluorescence intensity (MFI) of the markers (TPO: n = 12 MYH9-RD; n = 12 ANKRD26-RT; TPO +EPAG: n = 12 MYH9-RD; n = 12 ANKRD26-RT; p=NS). (F) Output was calculated as the fold increase in the percentage of CD41+CD42b+ cells in presence of TPO +EPAG with respect to the percentage of double-positive cells in presence of TPO alone (ANKRD26-RT: n = 12, p<0.05; MYH9-RD: n = 12, p<0.01). All results are presented as mean ± SD. The full data set is provided in Figure 4—source data 1.

Eltrombopag sustains increased proplatelet formation ex vivo.

(A) Confocal microscopy analysis of 3D megakaryocyte culture imaged at the end of differentiation. Megakaryocytes were elongating proplatelet shafts, which assembled nascent platelets at their terminal ends, within the hollow space of silk pores (red = CD61, blue = silk) (scale bars = 50 µm). (Av-viii) Analysis of proplatelet structure was performed by immunofluorescence staining of the megakaryocyte-specific cytoskeleton component β1-tubulin (red=β1-tubulin; blue = nuclei; scale bar = 25 µm). In both diseases, the representative pictures show increased elongation and branching of proplatelet shafts in presence of TPO +EPAG with respect to TPO alone. (B) The percentage of proplatelet forming megakaryocytes was calculated as the number of cells displaying long filamentous pseudopods with respect to the total number of round megakaryocytes per analyzed field (TPO: n = 12 MYH9-RD; n = 12 ANKRD26-RT; TPO +EPAG: n = 12 MYH9-RD; n = 12 ANKRD26-RT; **p<0.01; *p<0.05). All results are presented as mean ± SD. The full data set is provided in Figure 5—source data 1.

Ex vivo platelet count for predicting response to treatments.

(A) The flow chamber was perfused with culture media and released platelets collected into gas-permeable bags before counting by flow cytometry. (B) Light microscopy and immunofluorescent analysis of the collected medium demonstrated the presence of large pre-platelets, dumbbells, and little discoid platelets having the microtubule coil typically present in resting platelets (red=β1-tubulin, scale bars = 10 µm). (C) Representative flow cytometry analysis of expression of CD41 and CD42b surface markers. (D) Analysis of the correlation between the increase of platelet count analyzed ex vivo and the increase of platelet count observed in vivo from the same patients. For the ex vivo analysis, platelet count was calculated by flow cytometry with counting beads (n = 8 MYH9-RD; n = 9 ANKRD26-RT). (E) Analysis of the correlation between ex vivo megakaryocyte output and the increase of platelet count observed in vivo from the same patients. (n = 8 MYH9-RD; n = 9 ANKRD26-RT). The full data set is provided in Figure 6—source data 1.

Figure 6—source data 1

Analysis of platelet count and megakaryocyte output ex vivo, and correlation with platelet count in vivo.

https://cdn.elifesciences.org/articles/58775/elife-58775-fig6-data1-v1.xlsx
Figure 7 with 4 supplements
Assessment of iPSC megakaryocyte differentiation.

(A) iPSC clones were cultured for 18 days and analyzed by flow cytometry to assess megakaryocytes differentiation. Histograms show the mean fluorescence intensity (MFI) of MYH9-RD clones for CD42a, CD42b, CD41, and CD61 markers, relative to healthy controls (n = 3 Healthy Controls, n = 3 MYH9-RD; p=NS). (B) Representative images of proplatelet forming-megakaryocytes at day 19 of culture from different iPSC clones. (C) Percentage of proplatelet formation from the different genotypes (n = 6 Healthy Control; n = 6 MYH9-RD (each clone repeated two times); *p<0.05). (D) The number of bifurcation per single megakaryocyte from the different genotypes (*p<0.01). All results are presented as mean ± SD. The full data set is provided in Figure 7—source data 1. Assessment of iPSC clone pluripotency is shown in Figure 7—figure supplement 1. Morphological and genomic characterization of iPSC clones is shown in Figure 7—figure supplement 2. iPSCs haematopoietic differentiation is shown in Figure 7—figure supplement 3. Embryoid Bodies and trilineage differentiation of iPSC clones is shown in Figure 7—figure supplement 4.

Figure 7—source data 1

Analysis of iPSC differentiation and proplatelet formation.

https://cdn.elifesciences.org/articles/58775/elife-58775-fig7-data1-v1.xlsx
Figure 7—figure supplement 1
Characterization of iPSCs.

(A) Quantitative RT-PCR analysis of SOX2, NANOG, and OCT4 mRNA expression levels in healthy control and MYH9-RD clones. Expression levels were normalized relative to hESCs RC17. A control iPSC cell line (CTR2#6) was used as internal control. Results are presented as mean ± SEM (n = 2/clone; *p<0.05; **p<0.01). (B) Representative immunofluorescence staining of pluripotency markers OCT4, SOX2, NANOG, SSEA-4, and Tra-1–81.

Figure 7—figure supplement 1—source data 1

Analysis of gene expression in iPSCs.

https://cdn.elifesciences.org/articles/58775/elife-58775-fig7-figsupp1-data1-v1.xlsx
Figure 7—figure supplement 2
Morphological and genomic characterization of iPSC clones.

(A) Representative phase-contrast images of healthy controls and mutated clones. Magnification ×10. (B) Representative karyotype analysis showed a normal karyotype (healthy controls: upper panel, 46,XX; MYH9-RD bottom panel, 46,XY).

Figure 7—figure supplement 3
iPSCs hematopoietic differentiation.

Representative phase-contrast microscope images of iPSC colonies at day 0, hematopoietic progenitor.

Figure 7—figure supplement 4
Embryoid Bodies and trilineage differentiation of iPSC clones.

Phase: day 4 embryoid body culture (10 x). After 14 days, differentiated cultures exhibited the presence of cells immune-positive for endodermal (a–FP), mesodermal (a-SMA), and ectodermal (bIII-Tubulin), germ-layer markers. Magnification: ×10.

Validation of the system with iPSC mutated clones.

(A) Megakaryocytes were differentiated from iPSCs of patients affected by MYH9-RD and cultured for 14 days in a petri dish before passing into the bone marrow device in presence of 10 ng/mL TPO supplemented or not with 500 ng/mL EPAG. (B) Representative immunofluorescence staining of CD61 megakaryocytes (i,ii) and confocal microscopy analysis (iii-vi) of 3D megakaryocyte culture imaged at the end of differentiation. Megakaryocytes are elongating proplatelet shafts, which assemble nascent platelets at their terminal ends, within the hollow space of silk pores (red = CD61, blue = silk, scale bars = 50 µm). (C) Representative flow cytometry analysis of CD41+CD42b+ megakaryocytes at the end of the culture and (D) statistical analysis of mean fluorescence intensity (MFI) of the markers (n = 3 TPO, n = 3 TPO +EPAG; p=NS). (E) Output was calculated as the number of CD41+CD42b+ cells in presence of TPO +EPAG with respect to the percentage of double-positive cells in presence of TPO alone (n = 3 TPO, n = 3 TPO +EPAG; *p<0.05). (F) Platelet number was calculated by counting beads after perfusing the chamber. The fold increase was calculated as the number of ex vivo platelet count in the presence of TPO +EPAG with respect to TPO alone (n = 3 TPO, n = 3 TPO +EPAG; *p<0.05). All results are presented as mean ± SD. The full data set is provided in Figure 8—source data 1.

Figure 8—source data 1

Analysis of iPSC differentiation and platelet production in the presence of EPAG.

https://cdn.elifesciences.org/articles/58775/elife-58775-fig8-data1-v1.xlsx
Summary of the proposed workflow.

After sampling, hematopoietic stem and progenitors cell from patients can be differentiated into primary megakaryocytes (MKs) or transformed into induced Pluripotent Stem Cells (iPSCs). iPSC are subjected to quality check, expansion and banking. Megakaryocytic progenitors differentiated either from primary stem cells or iPSCs are seeded within a 3D bone marrow tissue device, cultured in the presence of the tested drug, and analyzed. After perfusion, platelets are collected and counted in order to assess patient-specific response. The figure of the microscope and tubes was adapted from Servier Medical Art licensed under a Creative Commons Attribution 3.0 Unported License (https://smart.servier.com).

Tables

Table 1
Main features of the study population.
ANKRD26-RTMYH9-RD
Total samples, no.1212
M/F9/35/7
Age - mean [range], years46
[22-67]
48
[26-59]
Platelet count - mean [range] x109/L32
[9-75]
29
[5-69]
Table 2
Main features of the study population treated with Eltrombopag in vivo and ex vivo.
ANKRD26-RTMYH9-RD
Patients treated with Eltrombopag in vivo and ex vivo, no.67*
M/F6/32/6
Age - mean [range], years47
[22-67]
45
[31-59]
Platelet count at baseline IN VIVO mean [range], x109/L36
[12-75]
24
[5-69]
Increase of platelet count after Eltrombopag treatment IN VIVO‡ - mean [range], x109/L34
[7-64]
88
[5-231]
Platelet count EX VIVO – TPO mean [range], x1048.3
[6-13]
7.8
[5-12]
Increase of platelet count EX VIVO§ TPO + EPAG mean [range], x10424
[0–50]
36
[1-105]
  1. * of whom one patient repeated two times ex vivo.

     of whom three patients repeated two times ex vivo.

  2.  Increase of platelet count with Eltrombopag with respect to baseline.

    § Increase of platelet count with Eltrombopag with respect to the untreated counterpart (TPO only).

Table 3
Comparison of silk-bone marrow models for megakaryocyte culture.
Di Buduo et al., 2015Di Buduo et al., 2017Current Manuscript
Size of the cell-seeding well15 × 20 × 5 mm (1500 mm3)3.5 × 20 × 5 mm (350 mm3)2 × 15 × 3.5 mm (105 mm3)
No. of chambers that can be perfused in parallelMax. 2Max. 1>2 (up to at least 4)
Source of bloodHuman
Umbilical Cord Blood
Human
Umbilical Cord Blood
Human
Peripheral Blood
Type of hemopoietic stem and progenitor cellsCD34+CD34+- CD45+
- CD34+-derived iPSCs
Type of cells seededCD34+-derived megakaryocytesCD34+-derived megakaryocytes- CD45+-derived megakaryocyte progenitors
- iPSC-derived megakaryocyte progenitors
No. of cells seeded2.5 × 1054 × 1055 × 104
Time of the culture24 hr24 hr>7 days
Major applicationStudying mechanisms of thrombopoiesisProof of concept for scaling up platelet productionDrug Testing in individual patients
Key resources table
Reagent type
(species) or
resource
DesignationSource or
reference
IdentifiersAdditional
information
 Gene (human)ANKRD26 – ankyrin repeat domain 26GenBankTHC2, bA145E8.1OMIM 188000
 Gene (human)MYH9 – myosin heavy chain 9GenBankBDPLT6, DFNA17, EPSTS, FTNS, MATINS, MHA, NMHC-II-A, NMMHC-IIA, NMMHCAOMIM 155100
 Genetic reagent (human)STR
D21S11, D7S820, CSF1PO, TH01, D13S317, D16S539, vWA, TPOX, D5S818
Amelogenin
Promega GenePrintGenetic multi-locus human DNA profile
Genetic loci
Cell line authentication
 Cell line (human)Human embryonic stem cellISENET
Biobank
Embryonic stem cell line RCe021-A (RC-17) Rosalin cells, RRID:CVCL_L206Control embryonic stem cell (human)
 Cell line (human)iPSC generated from healthy controlThis paperCTR2#6Control iPSC
(human)
 Cell line (human)iPSC generated from healthy controlThis paperCPNControl iPSC (human)
 Cell line (human)iPSC generated from MYH9-RD patientThis paperMHPatient iPSC (human)
Transfected construct
(human)
Sindai virus
OCT4, KLF4, Sox2, cMyc
Life TechnologiesCytoTune-iPS Sendai ReprogrammingGeneration of iPSCs (human)
Biological sample (human)Peripheral Blood---
Commercial assay or kitCytoTune-iPS 2.0 Sendai Reprogramming KitInvitrogen/Thermo Fisher Scientific#A16517Generation of iPSCs (human)
Commercial assay or kitVenor GeMMinerva Biolabs#11–1250Mycoplasma detection
Commercial assay or kitQiagen DNA Mini KitQiagen#A31881
DNA extraction
Commercial assay or kitTruCountBecton - Dickinson#340334FACS Counting Beads
Commercial assay or kitSlide-A-Lyzer Dyalisis Cassettes, 3.5K MWCO, 12 mLThermo Scientific#66110Dyalisis
Commercial assay or kitMiniMACS Starting KitMiltenyi Biotec# 130-090-312Cell sorting
AntibodyAnti-Human CD61 (Mouse Monoclonal)Beckman Coulter#IM0540, RRID:AB_28891761:100
AntibodyAnti - CD45 Microbeads (Mouse Monoclonal)Miltenyi Biotec#130-045-801,
RRID:AB_2783001
20 μl / 107 cells
AntibodyAnti - CD61 Microbeads (Mouse Monoclonal)Miltenyi Biotec#130-051-101
RRID:AB_2889174
20 μl / 107 cells
AntibodyAlexa Fluor 594 (Goat Anti Mouse)ThermoFisher Scientific# A11005,
RRID:AB_2534073
1:500
AntibodyFITC anti-human CD41 Antibody (Mouse Monoclonal)BioLegend#303703,
RRID:AB_314373
5 μl
Megakaryocytic marker
AntibodyPE anti-human CD42b Antibody (Mouse Monoclonal)BioLegend#303905,
RRID:AB_314385
5 μl
Megakaryocytic marker
Antibodya-FP
(Mouse Monoclonal)
R and D system#MAB1369,
RRID:AB_2258005
1:50
Trilineage differentiation
Antibodya-SMA
(Mouse Monoclonal)
Millipore Sigma#A2547,
RRID:AB_476701
1:200
Trilineage differentiation
AntibodybIII-Tubulin
(Mouse Monoclonal)
Millipore Sigma#MAB1637,
RRID:AB_2210524
1:100
Trilineage differentiation
AntibodySOX2
(Rabbit Monoclonal)
Abcam,#Ab97959,
RRID:AB_2341193
1:300 Pluripotency markers
AntibodyNanog
(Mouse Monoclonal)
Millipore#MABD24,
RRID:AB_11203826
1:500
Pluripotency markers
AntibodySSEA4
(Mouse Monoclonal)
Millipore#MAB4304,
RRID:AB_177629
1:50
Pluripotency markers
AntibodyTra1-81
(Mouse Monoclonal)
Millipore#MAB4381C3,
RRID:AB_2889175
1:50
Pluripotency markers
Sequence-based reagentGenePrint 10 SystemPromega#B9510STR genotyping
Peptide, recombinant proteinRecombinant Human Thrombopoietin (TPO)Peprotech#300–18Cytokine
Peptide, recombinant proteinRecombinant Human interleukin-11 (IL-11)Peprotech#200–11Cytokine
Peptide, recombinant proteinRecombinant Human interleukin-6 (IL-6)Peprotech#200–6Cytokine
Peptide, recombinant proteinRecombinant Human Fibroblast Growth Factor (FGF)Peprotech#100-18BCytokine
Peptide, recombinant proteinRecombinant Fms-related tyrosine kinase three ligand (Flt3L)Peprotech#300–19Cytokine
Peptide, recombinant proteinRecombinant Human Stem Cell Factor (SCF)Peprotech#300–07Cytokine
Peptide, recombinant proteinRecombinant Human VEGF165Peprotech#100–20Cytokine
Chemical compound, drugCHIR 99021Tocris#4423Pharmacological Inhibitor
Chemical compound, drugLithium Bromide (LiBr)Sigma-Aldrich#213225Silk processing
Chemical compound, drugPenicillin – Streptomycin 100XEuroclone#EB3001DAntibiotics
Chemical compound, drugParaformaldehyde (PFA)Sigma - Aldrich#158127Fixation
Chemical compound, drugTriton
X-100
Sigma - Aldrich#X100Permeabilization
Chemical compound, drugEltrombopagNovartisN/ADrug Testing
Software algorithmGeneMapper Software version 4.0Applied Biosystem#4440915, RRID:SCR_014290Genotyping Analysis
OtherHuman FibronectinBecton Dickinson# 354008Silk functionalization
OtherPoly(lactic acid)FormFutura-Scaffolding
OtherPluronic F-127Sigma-Aldrich#P244325%
OtherPDMS (SYLGARD 184 Silicone Elastomer)Dow Corning# 1673921Scaffolding
OtherDulbecco’s Phosphate Buffered Saline (PBS)Euroclone#ECB4053LSaline buffer
OtherEssential 8
Flex media
Gibco/Thermo Fisher#A2858501Culturing media
OtherStemSpan SFEM MediumVoden#09650Culture medium
OtherE8 mediumGibco/Thermo Fisher Scientific#A1517001Culture medium
OtherL-Glutamine 100XEuroclone#ECB300D1%
OtherHoechst 33258Sigma - Aldrich#8614051:10000
OtherProLong Gold antifade reagentInvitrogen#P36980Microscopy
OtherCFSE Cell Division Tracker KitBiolegend#4238015 mM
OtherGeltrexGibco/Thermo Fisher Scientific#A1413202Matrix
OtherVTN-NGibco/Thermo Fisher Scientific#A14700Matrix
OtherKaryoMax ColcemidGibco - Sigma329749009Chromosome/metaphase banding Genomic stability
OtherQuinacrineSigma/MerckQ3251Chromosome/metaphase banding Genomic stability
OtheraCGHAgilent TechnologiesSurePrint G3 Human CGH Microarray 8 × 60KDNA Analytics software v.5CNVs
Table 4
STR-genotype of iPSCs and donor cells.

Numbers in each locus refer to the number of repeats in each allele.

Cell lineTH01D21S11D5S818D13S317D7S820D16S539CSF1POAMELvWATPOX
CPN9.3, 9.328,
33.2
12, 128,1110,1111,1110,12X,X16,168,8
MH7,732.2, 33.212,1311,139,1112,1211,11X,Y15,1611,11
MH-17,732.2, 33.212,1311,139,1112,1211,11X,Y15,1611,11
MH-117,732.2, 33.212,1311,139,1112,1211,11X,Y15,1611,11
Table 5
Molecular karyotype.

aCGH analysis was performed by SurePrint G3 Human CGH Microarray Kit 60K and revealed CNVs on chromosomes 2 and 3 in MH-11 cells. No genomic instability was observed in CPN cells.

Cell lineMolecular karyotype
CPNarr[hg18] (1–22,X)x2
MH-11arr[hg18] 2p16.1 (60,966,923–61,003,123)x6, 3q13.31(117,895,555-118,070,129)x7
Table 6
Primer sequences for qRT-PCR.
GeneForward primerReverse primer
SOX2GACAGAGCCCATTTTCTCCAAAATCCTGTCCTCCCATTCC
NANOGTATGCCTGTGATTTGTGGGCGTTTGCCTTTGGGACTGGTG
OCT4AGAGGATCACCCTGGGATATCGCCGGTTACAGAACCACAC
GAPDHTGTTCGACAGTCAGCCGCATTAAAAGCAGCCCTGGTGACC
Author response table 1
Blood 2015Biomaterials 2017Current Manuscript
Size of the cell-seeding well15x20x5 mm (1500mm3)3.5x20x5 mm (350mm3)2x15x3.5 mm (105mm3)
No. of chambers that can be perfused in parallelMax. 2Max. 1> 2 (up to at least 4)
Source of bloodHuman
Umbilical Cord Blood
Human
Umbilical Cord Blood
Human
Peripheral Blood
Type of haemopoietic stem and progenitor cellsCD34+CD34+
  • CD45+

  • CD34+-derived iPSCs

Type of cells seededCD34+-derived megakaryocytesCD34+-derived megakaryocytes
  • CD45+-derived megakaryocyte progenitors

  • iPSC-derived megakaryocyte progenitors

No. of cells seeded2.5x1054x1055x104
Time of the culture24 hours24 hours> 7 days
Major applicationStudying mechanisms of thrombopoiesisProducing a high number of plateletsDrug Testing in individual patients

Additional files

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Christian A Di Buduo
  2. Pierre-Alexandre Laurent
  3. Carlo Zaninetti
  4. Larissa Lordier
  5. Paolo M Soprano
  6. Aikaterini Ntai
  7. Serena Barozzi
  8. Alberto La Spada
  9. Ida Biunno
  10. Hana Raslova
  11. James B Bussel
  12. David L Kaplan
  13. Carlo L Balduini
  14. Alessandro Pecci
  15. Alessandra Balduini
(2021)
Miniaturized 3D bone marrow tissue model to assess response to Thrombopoietin-receptor agonists in patients
eLife 10:e58775.
https://doi.org/10.7554/eLife.58775