This disclosure generally relates to endothelial cells and methods of making and using such cells.
This application claims the benefit of priority under 35 U.S.C. § 119(e) to U.S. Application No. 63/318,556 filed on Mar. 10, 2022.
The endothelium of the liver is heterogenous and carries out many functions essential for organ homeostasis. The liver sinusoidal endothelial cells (LSECs) that line the sinusoids of the organ represent a critical component of this endothelium, as they perform many organ-specific functions including passive blood component transport through fenestrations and active scavenging of circulating biomolecules and pathogens. LSECs also secrete several functional proteins including a number of ‘angiocrines’ that regulate hepatic regeneration as well as coagulation factor VIII (FVIII) that is critical for proper hemostasis.
Given this diverse range of activities, LSECs are directly or indirectly involved in many liver diseases. Disruption of FVIII production or activity results in the bleeding disorder Hemophilia A, whereas alterations in other LSEC functions are observed in metabolic diseases such as NAFLD and NASH. LSEC functions also are acutely altered during drug-induced liver injury and in conditions of chronic liver damage such as cirrhosis and liver cancer. Given their central role in normal liver physiology and disease processes, LSECs represent an important target population for developing new therapeutic interventions to treat some of the most devastating forms of these diseases.
As access to human LSECs is limited, recent efforts have focused on generating these cells from mouse and human pluripotent stem cells (hPSCs). It previously was shown that hPSC-derived angioblasts that displayed a venous phenotype could generate LSECs more rapidly than arterial-like cells following transplantation into recipient NSG mice. These observations were consistent with lineage tracing studies in mice and zebrafish, showing that LSECs derive from the developing venous vasculature. Although these studies identified the vascular subtype from which LSECs originate, the early developmental steps leading to the generation of this lineage (e.g., mesoderm induction) are not well understood.
The importance of mesoderm induction to subsequent lineage commitment is highlighted by the findings from lineage tracing in mice, which revealed that cell fate decisions are made during the time of gastrulation. For example, in the heart, ventricular and atrial cardiomyocytes are generated from different subpopulations of Mesp1-expressing mesoderm that are specified at different times. Similarly, different human heart and blood cell lineages are specified at the mesoderm stage during hPSC differentiation.
This disclosure describes a protocol for the generation of human pluripotent stem cell (hPSC)-derived venous endothelial cells (VECs) that display robust potential to engraft the liver of either normal or Hemophilia ANSG recipient mice, where the cells then differentiated into functional liver sinusoidal endothelial cells (LSECs). The experimental design uses a novel competitive repopulation assay to show that the efficiency of LSEC engraftment is directly related to the subtype of mesoderm induced in the cultures. The work described herein demonstrates that mesoderm characterized by the co-expression of KDR and CD235a/b (i.e., having a phenotype of KDR+ CD235a/b+) induced through optimal levels of BMP and Activin signaling generates a VEC population that displays up to 50-fold greater engraftment potential than venous angioblasts derived from expression of KDR and the lack of expression of CD235a/b (i.e., having a phenotype of KDR+ CD235a/b-) mesoderm generated with the previously described protocol. With these engraftment efficiencies, nearly all (>80%) of the murine sinusoidal endothelium can be replaced with human LSECs. In addition to producing a VEC population with dramatically higher LSEC potential than previous protocols, the protocols described in this disclosure are approximately 10-fold more efficient, yielding about 1 VEC per input hPSC.
The levels of human vascular engraftment achieved with the approach described herein are the highest reported to date. In addition, the levels are therapeutically relevant as it is demonstrated herein that the LSECs produced after engraftment were able to produce sufficient levels of FVIII to reverse the bleeding phenotype in Hemophilia A recipients. With these efficiencies, the methods and compositions described herein provide commercial applications for the development of vascular cell therapies for Hemophilia A and other liver diseases, as well as for ischemic diseases that affect the heart and skeletal muscle.
In one aspect, methods of making mesodermal cell-derived venous endothelial cells (VECs) are provided. Such methods typically include providing human pluripotent stem cells (hPSCs) having a pluripotent phenotype indicative of human induced pluripotent stem cells (iPSCs) or human embryonic stem cells (hESCs); contacting the hPSCs with modulators of BMP and Activin signaling under appropriate conditions to produce mesodermal cells having a phenotype of KDR+ and CD235a/b+; and inducing the mesoderm cells having the phenotype of KDR+ and CD235a/b+ to differentiate into mesodermal cell-derived VECs.
In some embodiments, the contacting step is performed in embryoid bodies (EBs). In some embodiments, the contacting step is performed in a monolayer. In some embodiments, the inducing step is performed in embryoid bodies (EBs). In some embodiments, the inducing step is performed in monolayer adherent form.
Representative modulators of BMP signaling include, without limitation, BMP4, BMP2, or small molecule BMP signaling agonists (e.g., ventromorphins [PMID 28787124], ID1 or ID2 [PMID 23527084], chromenone 1 [PMID 35108017], SB 4 [CAS number 100874-08-6] or similar). Representative modulators of Activin signaling include, without limitation, Activin A, NODAL, or small molecule TGFbeta signaling agonists.
In some embodiments, the mesoderm cells also have a phenotype of PDGFRa+, CD56+ and APLNR+. In some embodiments, the mesodermal cell-derived VECs have a phenotype of CD34+, CD31+, CD73+, and CD184-. In some embodiments, the mesodermal cell-derived VECs also have a phenotype of NRSF2+, NRP2+, NT5E+ and EPHB4+.
In some embodiments, the inducing takes place in the presence of VEGF-A, VEGF-B, VEGF-C, VEGF-D or PIGF, or small molecule VEGFA signaling agonists. In some embodiments, the inducing takes place in the presence of bFGF/FGF2 or small molecule FGF signaling agonists.
In some embodiments, VECs engraft in vivo and mature to liver sinusoidal endothelial cells (LSECs). In some embodiments, the VECs differentiate into functional liver sinusoidal endothelial cell-like cells (LSEC-LCs) having a phenotype of CD31+, CD32+ and LYVE1+. In some embodiments, the VEC-derived-LSECs further have a phenotype of CD32B+, STAB2+ and FVIII+.
In some embodiments, the LSECs express FCGR2B, LYVE1, STAB2, F8, CD14, MRC1, CD36 and RAMP3 at amounts that are greater than VEC-derived non-LSEC endothelium with a phenotype of CD31+ CD32- LYVE1-. In some embodiments, the LSECs express PECAM1, VWF and CALCRL at amounts that are less than VEC-derived non-LSEC endothelium with a phenotype of CD31+, CD32- LYVE1-. In some embodiments, the mesoderm cells generate a population of VECs that exhibits at least a 20-fold greater (e.g., 30-fold, 40-fold, 50-fold greater) engraftment potential than venous angioblast cells derived from mesoderm cells having a phenotype of KDR+ and CD235a/b-.
In another aspect, methods of treating a subject suffering from a liver disease are provided. Such methods typically include administering / delivering the cells of any one of the preceding claims to the subject, wherein the cells efficiently engraft in the liver, thereby treating the subject suffering from the liver disease.
In some embodiments, the liver disease is Hemophilia A or other monogenic endothelial disease. In some embodiments, the liver disease is acute drug liver injury (e.g., Sinusoidal Obstruction Syndrome (human monocrotaline toxicity), or Acetaminophen overdose), chronic liver injury (e.g., NASH, NAFLD, Cirrhosis, chronic drug injury), liver cancer (e.g., primary, hepatocellular carcinoma; or secondary (e.g., colon, breast, pancreatic) liver metastatic cancer).
In some embodiments, the engrafted cells differentiate into LSECs. In some embodiments, the LSECs express therapeutic amounts of FVIII (e.g., sufficient levels of FVIII to reverse the severe bleeding phenotype in a subject suffering from Hemophilia A).
In some embodiments, the administering / delivering step is repeated more than once. In some embodiments, the administering / delivering step comprises intravenous delivery (e.g., via infusion via the hepatic portal vein or systemically).
In some embodiments, the hPSCs, mesoderm cells, VECs or LSECs are rendered hypoimmunogenic through genetic addition or removal of immunoregulatory antigens, resulting in cell populations that are not readily rejected by the recipient despite incomplete immunological matching. In some embodiments, administering / delivering functional LSEC progenitors slows or reverses disease progression.
In some embodiments, the hPSCs are genetically modified such that the VECs have designer or specialized new functions (e.g., where novel exogenous therapeutic proteins are expressed to provide novel therapeutic applications). The list of such exogenous proteins is virtually endless.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the methods and compositions of matter belong. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the methods and compositions of matter, suitable methods and materials are described below. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety.
Liver sinusoidal endothelial cells (LSECs) form the predominant microvasculature in the liver where they carry out many functions including the secretion of coagulation factor VIII (FVIII). To investigate the early origins of human LSECs, an efficient and scalable protocol was developed to produce human pluripotent stem cell (hPSC)-derived venous endothelium from different mesoderm subpopulations. Using a sensitive and quantitative vascular competitive transplantation assay, it was demonstrated that venous endothelial cell (VEC) populations generated from BMP4-induced and Activin A-induced mesoderm characterized by KDR+ CD235a/b+ expression was 50-fold more efficient at engrafting the LSEC compartment in the liver of NSG mice than venous populations generated from KDR+ CD235a/b-mesoderm induced by BMP4 and WNT agonism.
When transplanted into immunocompromised Hemophilia A mice (NSG-HA), these VECs engrafted the liver, proliferated, and generated functional LSECs that secreted bioactive FVIII capable of correcting the bleeding phenotype. Together, these findings highlight the importance of appropriate mesoderm induction for the generation of specific cell types from hPSCs and demonstrate that, with this approach, it is possible to generate functional LSECs in a pre-clinical model of Hemophilia A.
Thus, this disclosure describes methods of making mesodermal cell-derived VECs. As described herein, such mesodermal cell-derived VECs subsequently can be used to produce LSECs in vitro or in vivo, which have a number of therapeutic applications.
Human pluripotent stem cells (hPSCs) can be used in the methods described herein. Human pluripotent stem cells (hPSCs) include, without limitation, human embryonic stem cells (hESCs), human induced pluripotent stem cells (iPSCs) or other human cells having a pluripotent phenotype.
The hPSCs can be contacted with at least one modulator of BMP signaling and at least one modulator of Activin signaling in order to generate the mesodermal cells described herein (i.e., KDR+, CD235a/b+). Modulators of BMP signaling and modulators of Activin signaling are known in the art; representative modulators of BMP signaling include, without limitation, BMP4, BMP2, or small molecule BMP signaling agonists (e.g., ventromorphins [PMID 28787124], ID1 or ID2 [PMID 23527084], chromenone 1 [PMID 35108017], SB 4 [CAS number 100874-08-6] or similar), while representative modulators of Activin signaling include, without limitation, Activin A, NODAL, or small molecule TGFbeta signaling agonists.
As described herein, under appropriate conditions, mesoderm cells having a phenotype of KDR+ and CD235a/b+ are produced. The mesodermal KDR+ CD235a/b+ cells also can be phenotypically PDGFRa+, CD56+ and APLNR+.
Also as described herein, under appropriate conditions, the mesodermal KDR+ CD235a/b+ cells can be induced to differentiate into VECs (i.e., “mesodermal cell-derived VECs”), which typically have a phenotype of CD34+, CD31+, CD73+ and CD184-. The mesodermal cell-derived VECs also can be phenotypically NR2F2+, NRP2+, NT5E+ and EPHB4+. Any number of compounds can be used to induce the mesodermal KDR+ CD235a/b+ cells into mesodermal cell-derived VECs. For example, mesodermal cell-derived VECs cells can be induced using VEGF-A, VEGF-B, VEGF-C, VEGF-D, PIGF, bFGF/FGF2, or small molecule signaling agonists of VEGFA or FGF.
It would be appreciated that the contacting step and the inducing step can be performed in culture (e.g., monolayer, adherent) or in embryoid bodies (EBs). Culture conditions for maintaining or differentiating the cells are described herein, and embryoid bodies are known in the art as aggregates of pluripotent stem cells or derivative differentiating cells.
Significantly, the mesodermal cell-derived VECs described herein can be differentiated into liver sinusoidal endothelial cell-like cells (LSEC-LCs), which are phenotypically CD31+, CD32+ and LYVE1+. The LSEC-LCs also can be phenotypically CD32B+, STAB2+ and FVIII+. The differentiation into LSEC-LCs can take place in vitro (e.g., in culture), however, as described herein, the mesodermal cell-derived VECs differentiate extremely efficiently into functional LSECs in vivo. For example, the mesodermal cells described herein (i.e., KDR+ CD235a/b+) are capable of generating a population of VECs that exhibits at least a 20-fold greater (e.g., at least a 30-fold, 40-fold, 50-fold greater) engraftment potential than venous angioblast cells derived from mesoderm cells having a phenotype of KDR+ and CD235a/b-
Any number of compounds can be used to induce the mesodermal cell-derived VECs into LSEC-LCs in vitro. For example, mesodermal cell-derived VECs cells can be induced to form LSEC-LCs in the presence of bFGF, TGFbeta inhibition, cAMP agonism, and an agonist of BMP9 signaling. Alternatively, mesodermal cell-derived VECs can be induced to differentiate into LSEC-LCs using hypoxic conditions.
The LSECs described herein (i.e., derived from the engraftment of mesodermal cell-derived VECs) can be characterized by greater levels of expression of FCGR2B, LYVE1, STAB2, F8, CD14, MRC1, CD36 and RAMP3 and lesser levels of expression of PECAM1, VWF and CALCRL relative to VEC-derived non-LSEC endothelium (i.e., having a phenotype of CD31+ CD32- LYVE1-).
The cells described herein, particularly the mesodermal cell-derived VECs, can be used to treat a subject suffering from a liver disease. For example, the cells described herein can be administered or delivered to the subject, resulting in efficient engraftment in the liver. As used herein, treating refers to an approach intended to obtain a beneficial or desired result, which may include alleviation of symptoms, or delaying or ameliorating a disease progression. It would be appreciated that cells can be administered to a subject via intravenous delivery (e.g., infusion via the hepatic portal vein or systemic circulation). In some instances, it may be desirable to administer or deliver cells more than once to a subject (e.g., multiple times, a plurality of times).
As demonstrated herein, engrafted mesodermal cell-derived VECs efficiently differentiate into LSECs and are able to produce therapeutically effective amounts of the Factor VIII (FVIII) protein (e.g., sufficient levels to reverse the severe bleeding phenotype in a subject suffering from Hemophilia A), thereby treating Hemophilia A. Given the number of cells that engraft and differentiate, however, any number of other monogenic endothelial diseases can be treated in addition to other liver diseases such as, without limitation, acute drug liver injury (e.g., Sinusoidal Obstruction Syndrome (e.g., human monocrotaline toxicity among other agents), or Acetaminophen overdose), chronic liver injury (e.g., NASH, NAFLD, Cirrhosis, chronic drug injury), liver cancer (e.g., primary, hepatocellular carcinoma; or secondary (e.g., colon, breast, pancreatic) liver metastatic cancer).
In some instances, it may be desirable to render the hPSCs, the mesoderm cells, the mesodermal cell-derived VECs or the LSECs hypoimmunogenic. Cells can be rendered hypoimmunogenic by genetic addition or removal of immunoregulatory antigens, thereby resulting in cell populations that are not readily rejected by the recipient despite incomplete immunological matching.
Similarly, in some instances, it may be desirable to genetically engineer the hPSCs, the mesoderm cells, the mesodermal cell-derived VECs or the LSECs. Any of such cells can be genetically engineered to express nucleic acid encoding one or more exogenous proteins (e.g., functional, structural and/or therapeutic).
In accordance with the present invention, there may be employed conventional molecular biology, microbiology, biochemical, and recombinant DNA techniques within the skill of the art. Such techniques are explained fully in the literature. The invention will be further described in the following examples, which do not limit the scope of the methods and compositions of matter described in the claims.
All experiments were done in accordance with institutional guidelines approved by the University Health Network Animal care committee for studies utilizing NSG mice in Toronto Canada (NOD.Cg PrkdcscidI12rgtm1Wjl /SzJ; Jackson Laboratory, local Ontario Cancer Institute colony). Experiments involving previously described NSG-HA mice (Merlin et al., 2019, Blood Adv., 3:825-38; Zanolini et al., 2015, Haematologica, 100:881-92) were performed at the Università del Piemonte Orientale following institutional approval. All animal studies were performed in 5-15 week old male and female mice for adult studies and P2-3 NSG neonates. Animals were maintained under standard conditions (12-hour light/dark cycle) in ventilated sterile microisolation cages as previously described (Gage et al., 2020, Cell Stem Cell, 27:254-69).
hPSC lines were maintained on gelatin (0.1% w/v in PBS) and growth factor reduced Matrigel (0.25%v/v, Corning) coated dishes with irradiated mouse embryonic fibroblasts in DMEM/F12 KnockOut serum based hPSC culture media as previously described (Gage et al., 2020, Cell Stem Cell, 27:254-69) with 10-20 ng/ml rhbFGF (R&D). All cell lines were authenticated by providence, fluorescent protein expression, and confirmed to be karyotypically normal and mycoplasma free within two passages of experimental use. HES2 hESCs (Reubinoff et al., 2000, Stem Cells Transl. Med., 9:686-96) (karyotype: 46XX) were previously targeted at the human ROSA locus to constitutively express tdRFP or eGFP (Irion et al., 2007, Nat. Biotechnol., 25:1477-82). H1-GFP hESCs (karyotype: 46XY) were a gift from R. Moon (University of Washington, Seattle, USA) (Davidson et al., 2012, PNAS USA, 109:4485-90). PGPC17-11 iPSCs (karyotype: 46XY) (Hildebrandt et al., 2019, Stem Cell Reports, 13:1126-41) obtained from J. Ellis (SickKids Research Institute, Toronto, Ontario, Canada) were adapted from culture in mTeSR1 (Stemcell Technologies) to culture with irradiated mouse embryonic fibroblasts. These iPSCs were maintained as above with the addition of ROCK inhibitor (Y-27623, 10 µM, TOCRIS) to cultures at the time of passage and EB formation.
hESC differentiation (
On day 4, mesoderm specified EBs were dissociated (Trypsin-EDTA, Corning, 5 min) stopped with 50% v/v fetal calf serum (FCS, Wiscent) / IMDM, counted and replated on GFR-Matrigel coated (2.5% v/v in IMDM) 6-well or 10 cm plates to specify venous endothelium. 1×106 cells / 6-well or 6×106 cells / 10-cm plate were plated in “base media” supplemented with rhbFGF (30 ng/ml) and VEGF-A (10 ng/ml, R&D). On day 6, media was removed and replaced with freshly prepared day 4 media. To specify arterial lineage cells, day 4-8 media consisted of “base media” supplemented with rhbFGF (30 ng/ml) and VEGF-A (100 ng/ml). On day 8, endothelial cells were isolated by CD34 targeted MACS before cryopreservation (Cryostore CS10, STEMCELL Technologies) for future studies. Monolayer cultures were washed (PBS -Mg-Ca), dissociated (TrypLE, 10 mins, 37° C.), and stained for CD34 MACS (Miltenyi, 130-146-702) using 10 µl antibody / 5×106 cells / 100 µl in base media supplemented with DNASE (1 U/ml, Millipore) for 30 minutes at 4° C. before purification over two columns in series (either MS or LS depending on cell number) to isolate populations consisting of 95% CD34+ cells or greater.
For competitive transplantation studies (
In vitro derived single cell suspensions from day 4 EBs (Trypsin-EDTA, 5 min, FCS stopped), day 8 monolayers (PBS washed, TrypLE, 10 min), day 8 EBs (Trypsin-EDTA, 5 min, 0.2% w/v Collagenase Type 1, 60 mins, 37° C.) were used for flow cytometric staining using the following pre-conjugated antibodies: anti-KDR-biotin (R&D, Clone 89106, 15:100), Streptavidin-PeCy7 (BD Biosciences, 1:100) Streptavidin-BV421 (BioLegend, 1:100), anti-CD235a/b-APC (BD Biosciences, Clone GA-R2/HIR2, 2:100), anti-CD34-PeCy7 (EBiosciences, Clone 4H11, 1:100), anti-CD31-APCCy7 (BD Biosciences, Clone WM59, 1:100), anti-CD31-FITC (BD Biosciences, Clone WM59, 15:100) anti-CD184-BV421 (BioLegend, Clone 12G5, 1:100), anti-CD73-APC (BD Biosciences, Clone AD2, 0.25:100). In vitro surface marker staining for 20-30 minutes at 4° C. in FACS buffer (PBS without Mg/Ca with 5% FCS (Wisent), 0.02% NaN3 (SigmaAldrich), DNASE (1 U/ml)). Following liver dissociation and non-parenchymal cell recovery, cells (1×106 cells / 100 µl) were treated with FcR blocking human IgG (Miltenyi, 15:100) in RPMI1640 with 0.5% BSA and 1 U/ml DNAse along with primary and secondary antibodies: anti-CD31-APCCy7 (BD Biosciences, Clone WM59, 1:100), rabbit-antiLYVE1 (Abcam, Polyclonal, 1:1000), and anti-CD32-PeCy7 (BioLegend, Clone FUN-2, 3:100), Donkey-anti-rabbit-APC (F′(ab)2 fragment, Jackson, 0.5:100). For cell sorting and engraftment analysis, samples were stained in IMDM with 0.5% FCS, DNASE (0.1 U/ml), and DAPI (Biotium, 0.1 µg/ml) before analysis using a LSRII flow cytometer (BD) or a Fortessa flow cytometer equipped with a 532 nm green laser (BD) or sorting with a FACSAriaIII (BD) instrument (532 nm laser) at the SickKids/UHN flow cytometry facility. Data were analyzed using FlowJo software (version 10.6.1, Tree Star).
Total RNA was isolated from bulk, MACS-enriched, or FACS-purified in vitro or in vivo hPSC-derived samples using RNAqueous-micro kit (Invitrogen) with post column DNAse treatment. cDNA synthesis (iSCRIPT, BioRad) was performed using up to 1 µg of RNA subsequent RT-qPCR was performed using a CFX384 Touch Real-Time instrument (BioRad), QuantiFast SYBR Green PCR kit (Qiagen) or SsoAdvanced Universal SYBR® Green Supermix (BioRad) and primers described in Table 1, following manufacture recommendations. Gene expression relative to TBP was determined from technical duplicates evaluated for relative copy number, reaction efficiency, and genomic DNA contamination (<0.01% of TBP content) using a 10-fold dilution series of sonicated human genomic DNA standards made in house from wild type HES2 hESC cells ranging from 2.5 pg/µl to 25 ng/µl. Heatmaps and bar graphs of gene expression were generated in Prism version 9 (Graphpad) with the exception of
Cell transplantation from previously frozen day 8 cells was performed as described using an irradiated neonate or monocrotaline (MCT, SigmaAldrich) conditioned adult NSG models (Gage et al., 2020, Cell Stem Cell, 27:254-69). In the neonatal model, NSG P1-P4 pups were irradiated (100 cGy) 18-24 hours before direct intrahepatic injection of 1.5×106 cells in 30 µl of base media by 30 G½ needle on a Hamilton syringe. In the adult model, 8-20 week old male and female NSG mice received a 150 mg/kg intraperitoneal MCT injection 18-24 hours before surgical cell delivery via intrasplenic injection of 1.5×106 cells in 40-50 µl of base media. After cell therapy, mice were transitioned to enrofloxacin for 14 days and grafts were allowed to expand and mature for up to 171 days.
Competitive cell transplantation experiments were performed in MCT-conditioned adult male NSG mice. Cell mixtures were prepared from day 8 CD34+ venous cell populations with similar post-thaw viability above 85% DAPI-. On the day of transplantation, cells were thawed, counted and mixed in indicated defined ratios such that the total inoculum contained 1×106 cells (for example 5×105 GFP and 5×105 RFP). Each inoculum was delivered to n=2-6 replicate mice via intrasplenic surgical delivery followed by 40-45 days of in vivo graft expansion and maturation.
Graft composition analysis from neonatal, adult or competitive adult NSG transplantation experiments was achieved by liver dissociation and non-parenchymal cell (NPC) isolation at the same time as heparinized cardiac blood sampling and histological sampling as previously described in detail (Gage et al., 2020, Cell Stem Cell, 27:254-69). Briefly, engrafted livers were mechanically and enzymatically dissociated to single cells by 40 minutes rotating in a 10 cm plate at 70 RPM (37° C., 5% CO2, 95% Air) in 24 ml liver dissociation solution comprising (66% v/v HANKS buffer, 33% v/v RPMI1640, BSA (0.17% w/v, Sigma-Aldrich), collagenase type 1 (0.3125% w/v, Sigma-Aldrich) and DNAse (1 U/ml). NPC liver fraction, lysed of red blood cells was sampled for flow cytometric graft composition assessment with or without FACS mediated cell recovery for RT-qPCR analysis.
Cell transplantation in NSG-HA mice was performed as described (Follenzi et al., 2008, J. Clin. Invest., 118:935-45; Olgasi et al., 2018, Mol. Ther. Methods Clin. Dev., 23:551-66). NSG-HA mice were treated with MCT (150 mg/kg) 18-24 hours before surgery. On the day of surgery, NSG-HA mice received 4 U of rhFVIII (Kovaltry, Bayer) intravenously to temporarily facilitate coagulation for surgery after which 1.5×106 (cohort 1, HES2-GFP) or 3.0×106 (cohort 2, HES2-RFP) day 8 CD34+ venous ECs derived from KDR+CD235a/b+ day 4 populations were delivered via intraportal or intrasplenic surgical delivery routes. Cohort 1 mice were terminated at 12 weeks post cell delivery and Cohort 2 mice were terminated at 16 weeks post cell delivery for FVIII antigen and histological analysis.
Functional FVIII activity in control and cell treated mice was assessed by activated partial thromboplastin time (aPTT) and tail clip bleeding time assays as previously described (Merlin et al., 2019, Blood Adv., 3:825-38). For longitudinal tracking aPTT assays, 3.2% citrated peripheral blood derived plasma (2000×g, 15 min) was compared to standard curves of serially diluted full length recombinant FVIII (Kovaltry) in pooled hemophiliac mouse plasma. Results are expressed as IU/ml where 1 IU/ml is 100% normal human FVIII activity benchmarked to the WHO international standard. For blood loss assays, the distal 2-2.5 mm of tail was cut off from anesthetized mice and blood was collected for 10 minutes in 14 ml of warmed (37° C.) saline with bleeding times recorded. Precipitated RBC containing blood cells were lysed and absorbance was measured at 575 nm, using a VictorX spectrophotometer (Perkin Elmer). Human specific FVIII antigen levels in NSG, and NSG-HA mice were determined by ELISA (Affinity Biologicals, FVIII-AG, Lots: AG80050 and AG8-0055) using heparinized cardiac puncture samples as previously described (Gage et al., 2020, Cell Stem Cell, 27:254-69) following manufacture recommended protocols with 1:1-1:2 diluted samples to reduce matrix effects. FVIII antigen levels are reported as IU/ml where 1 IU/ml represents 100% normal human antigen levels benchmarked to the WHO international standard.
At various time points after transplantation (10-24 weeks), liver tissue samples were fixed in PFA (4%w/v, 24-48 hours, 4° C., Electron Microscopy Services), stored in ethanol (70% v/v, 1-30 days, Commercial Alcohols) followed by paraffin processing and immunohistochemical staining by the UHN Pathology Research Program as previously described (Gage et al., 2020, Cell Stem Cell, 27:254-69). Primary antibodies included: rabbit anti Ku80 (Cell Signaling, 2180, clone C48E7, 1/1000 overnight), rabbit anti human LYVE1 (Abcam, Ab36993, polyclonal, 1/1000 overnight), mouse anti human CD31 (DAKO, M0823, clone JC/70A, 1/50 overnight), goat anti human CD32B (NSJ Bioreagents, R34966, polyclonal, 1/1000 overnight), sheep anti human Stabilin 2 (R&D, AF3645, polyclonal, 1/1000 overnight), goat anti GFP (Rockland, 600-101-215, polyclonal, 2/100 overnight). Secondary kits and developing reagents, MACH4 (Inter Medico, BC-M4U534L), IMMPRESS-AP, (Vector, MP5401-15), developers (Inter Medico, BC-WR806H or DAKO, K3468) were used as per kit instructions. Sections were imaged at the Advanced Optical Microscopy Facility using a Scan-Scope AT2 (Aperio) slide scanner, 40x objective and associated software. Immunofluorescent confocal microscopy was performed on 4-5 µm thick liver sections following sodium citrate (pH 6.0) antigen retrieval and casein blocking (Dako). Overnight applied primary antibodies included: rabbit anti Ku80 (Cell Signaling, 2180, clone C48E7, 1/100), mouse anti FVIII (Abcam, AB41188, clone 27.4, 2/100), goat anti GFP (Rockland, 600-101-215, polyclonal, 2/100), mouse anti Ki67 (Dako, M7240, clone MIB-1, 1/100), rabbit anti SMA (Abcam, Ab32575, clone E184, 1/100), mouse anti Vimentin (Sigma-Aldrich, V6630, clone V9, 1/100), rabbit anti Collagen I (Abcam, Ab34710, polyclonal, 1/100), and rabbit anti Collagen III (Abcam, Ab7778, polyclonal, 1/100). Secondary antibodies (Invitrogen) including donkey anti host - AF488, AF555, or AF647 variants applied for 1-2 hours at room temperature with DAPI counterstain followed by washing and mounting (Prolong Diamond Antifade, Invitrogen). Immunofluorescent slides were imaged using a Leica SP8 confocal imaging system (405 nm, 488 nm, 552 nm, and 638 nm laser lines) through a HC PL APO CS2 63x / 1.40 NA oil objective lens and images were processed for presentation using ImageJ (FIJI) (Schindelin et al., 2012, Nat. Methods, 9:676-82).
All data are presented as mean ± standard error of the mean (SEM) with sample sizes (n) depicted as individual dots on bar graphs or specified in figure legends and represent biological replicates as either independent cell differentiations or mice. Different “Cohorts” use independent in vitro differentiation biological replicates. No statistical method was used to pre-determine the sample sizes and neither randomization nor investigator blinding was performed on samples. Statistical significance was determined in Prism 9 (Graphpad) software using students T-test, one-way ANOVA, or two-way ANOVA analysis with Bonferroni post-hoc test as indicated. Results were considered to be significant at p < 0.05 (*/#), p < 0.01 (**/##), p < 0.001 (***/###) with specific comparisons indicated in the respective figure legend.
As we have shown in earlier studies that KDR+ mesoderm subpopulations separated based on the co-expression of CD235a/b display different hematopoietic and cardiovascular potential, we were interested in determining if LSEC potential would also differ between them. At the same time, we also aimed to improve the yield of venous endothelial cells by simplifying our endothelial specification to a monolayer format free from exogenous Notch inhibition. For these studies, hPSCs were differentiated using our well established embryoid body (EB)-based protocol that involves the formation of EBs (day 0-1), the induction of a primitive streak population (PS) and mesoderm (days 1-4) and the specification of mesoderm to an endothelial fate (days 4-8) (
To measure the venous endothelial potential of these mesoderm populations, we next dissociated the EBs at day 4 and re-plated the cells in a monolayer culture on Matrigel coated plates in a simplified venous endothelial specification media (10 ng/ml VEGF-A, 30 ng/ml bFGF) for 4 days. Total cell numbers were similar in all conditions (
To optimize production of the target VEC population, we translated the protocol from 6-well plates to 10 cm dishes, a change that yielded 7-fold higher mesoderm, 5-fold high total cell numbers (day 8) and 11-fold more CD34+ cells (day 8) compared to our previously published venous angioblast production protocol (Gage et al., 2020, Cell Stem Cell, 27:254-69) (
To evaluate the potential of the day 8 VEC populations to generate mature functional LSECs, we transplanted them into either irradiated neonatal NGS pups or into monocrotaline (MCT) conditioned NSG adults as described (Gage et al., 2020, Cell Stem Cell, 27:254-69) (
To establish the kinetics of engraftment, we analyzed transplanted mice at weekly intervals over the first 21 days and then at day 100. A low frequency of hPSC-derived GFP+ cells (0.14±0.02% of NPC) was detected one day following transplantation. This frequency increased to 2.19±0.62% by day 21 and to 38.6±1.5% by day 100 (
To ensure that this protocol is adaptable to different hPSC lines, we applied it to the H1-GFP hESC line (
The levels of engraftment (60% of NPC) achieved in this study are higher than the highest levels we reported previously (48±4%) (Gage et al., 2020, Cell Stem Cell, 27:254-69) suggesting that the day 8 VEC population produced with our modified protocol has greater LSEC potential than that generated with our original protocol. To be able to quantify LSEC engraftment, we established a competitive repopulation assay, similar to that used to quantify hematopoietic stem cell engraftment (Harrison, 1980, Blood, 55:77-81) (
Using this competitive assay, we next compared the engraftment potential of ECs generated with our new and old protocols. For this comparison, we transplanted equal mixtures (50:50) of our previously described venous angioblasts (RFP) and our K+2+derived VECs (GFP) (
As we made two major changes in our protocol, mesoderm induction and endothelial specification, we were next interested in determining which contributed to the improved engraftment potential. To address this, we tested the two stages of differentiation independently using the following experimental design. In the first arm, mesoderm induced with the two different protocols was specified to an endothelial fate using the monolayer strategy described in this study, while in the second arm, mesoderm induced with the new protocol was specified using either the EB approach of our earlier study or the monolayer format (
To determine if the improved engraftment potential of the day 4 12B4A-induced mesoderm was restricted to the K+2+ fraction, we next competed VECs generated from K+2- mesoderm generated from RFP-hPSCs to K+2+ generated from GFP-hPSCs (
To formally test the feasibility of treating FVIII-deficient Hemophilia A with cell replacement therapy, we transplanted Hemophilia A NSG mice that lack FVIII activity (NSG-HA) (Zanolini et al., 2015, Haematologica, 100:881-92) with the day 8 hPSC-derived VECs. Two cohorts of animals were transplanted by direct injection either into the portal vein (intraportal, IP) or into the spleen (intrasplenic, IS). Analyses of the first cohort (1.5 × 106 cells per mouse) over the 2-12 week monitoring period showed that the transplanted animals had human FVIII activity as measured by one-stage aPTT assay (0.011-0.035 IU/m1, 1.1-3.5% of normal human levels) standardized to Kovaltry (recombinant full length human FVIII) (
It is to be understood that, while the methods and compositions of matter have been described herein in conjunction with a number of different aspects, the foregoing description of the various aspects is intended to illustrate and not limit the scope of the methods and compositions of matter. Other aspects, advantages, and modifications are within the scope of the following claims.
Disclosed are methods and compositions that can be used for, can be used in conjunction with, can be used in preparation for, or are products of the disclosed methods and compositions. These and other materials are disclosed herein, and it is understood that combinations, subsets, interactions, groups, etc. of these methods and compositions are disclosed. That is, while specific reference to each various individual and collective combinations and permutations of these compositions and methods may not be explicitly disclosed, each is specifically contemplated and described herein. For example, if a particular composition of matter or a particular method is disclosed and discussed and a number of compositions or methods are discussed, each and every combination and permutation of the compositions and the methods are specifically contemplated unless specifically indicated to the contrary. Likewise, any subset or combination of these is also specifically contemplated and disclosed.
Number | Date | Country | |
---|---|---|---|
63318556 | Mar 2022 | US |