Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Cell ; 151(3): 559-75, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-23084400

ABSTRACT

ETS transcription factors ETV2, FLI1, and ERG1 specify pluripotent stem cells into induced vascular endothelial cells (iVECs). However, iVECs are unstable and drift toward nonvascular cells. We show that human midgestation c-Kit(-) lineage-committed amniotic cells (ACs) can be reprogrammed into vascular endothelial cells (rAC-VECs) without transitioning through a pluripotent state. Transient ETV2 expression in ACs generates immature rAC-VECs, whereas coexpression with FLI1/ERG1 endows rAC-VECs with a vascular repertoire and morphology matching mature endothelial cells (ECs). Brief TGFß-inhibition functionalizes VEGFR2 signaling, augmenting specification of ACs into rAC-VECs. Genome-wide transcriptional analyses showed that rAC-VECs are similar to adult ECs in which vascular-specific genes are expressed and nonvascular genes are silenced. Functionally, rAC-VECs form stable vasculature in Matrigel plugs and regenerating livers. Therefore, short-term ETV2 expression and TGFß inhibition with constitutive ERG1/FLI1 coexpression reprogram mature ACs into durable rAC-VECs with clinical-scale expansion potential. Banking of HLA-typed rAC-VECs establishes a vascular inventory for treatment of diverse disorders.


Subject(s)
Amniotic Fluid/cytology , Cell Differentiation , Endothelial Cells/cytology , Proto-Oncogene Proteins c-ets/metabolism , Retroviridae Proteins, Oncogenic/metabolism , Transforming Growth Factor beta/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans
2.
Blood ; 141(5): 503-518, 2023 02 02.
Article in English | MEDLINE | ID: mdl-35981563

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive and often incurable disease. To uncover therapeutic vulnerabilities, we first developed T-ALL patient-derived tumor xenografts (PDXs) and exposed PDX cells to a library of 433 clinical-stage compounds in vitro. We identified 39 broadly active drugs with antileukemia activity. Because endothelial cells (ECs) can alter drug responses in T-ALL, we developed an EC/T-ALL coculture system. We found that ECs provide protumorigenic signals and mitigate drug responses in T-ALL PDXs. Whereas ECs broadly rescued several compounds in most models, for some drugs the rescue was restricted to individual PDXs, suggesting unique crosstalk interactions and/or intrinsic tumor features. Mechanistically, cocultured T-ALL cells and ECs underwent bidirectional transcriptomic changes at the single-cell level, highlighting distinct "education signatures." These changes were linked to bidirectional regulation of multiple pathways in T-ALL cells as well as in ECs. Remarkably, in vitro EC-educated T-ALL cells transcriptionally mirrored ex vivo splenic T-ALL at single-cell resolution. Last, 5 effective drugs from the 2 drug screenings were tested in vivo and shown to effectively delay tumor growth and dissemination thus prolonging overall survival. In sum, we developed a T-ALL/EC platform that elucidated leukemia-microenvironment interactions and identified effective compounds and therapeutic vulnerabilities.


Subject(s)
Endothelial Cells , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Humans , Endothelial Cells/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Cell Communication , Coculture Techniques , Tumor Microenvironment
3.
Nature ; 545(7655): 439-445, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28514438

ABSTRACT

Developmental pathways that orchestrate the fleeting transition of endothelial cells into haematopoietic stem cells remain undefined. Here we demonstrate a tractable approach for fully reprogramming adult mouse endothelial cells to haematopoietic stem cells (rEC-HSCs) through transient expression of the transcription-factor-encoding genes Fosb, Gfi1, Runx1, and Spi1 (collectively denoted hereafter as FGRS) and vascular-niche-derived angiocrine factors. The induction phase (days 0-8) of conversion is initiated by expression of FGRS in mature endothelial cells, which results in endogenous Runx1 expression. During the specification phase (days 8-20), RUNX1+ FGRS-transduced endothelial cells commit to a haematopoietic fate, yielding rEC-HSCs that no longer require FGRS expression. The vascular niche drives a robust self-renewal and expansion phase of rEC-HSCs (days 20-28). rEC-HSCs have a transcriptome and long-term self-renewal capacity similar to those of adult haematopoietic stem cells, and can be used for clonal engraftment and serial primary and secondary multi-lineage reconstitution, including antigen-dependent adaptive immune function. Inhibition of TGFß and CXCR7 or activation of BMP and CXCR4 signalling enhanced generation of rEC-HSCs. Pluripotency-independent conversion of endothelial cells into autologous authentic engraftable haematopoietic stem cells could aid treatment of haematological disorders.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Endothelium/cytology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Adaptive Immunity , Aging/genetics , Animals , Cell Line , Cell Lineage , Cell Self Renewal , Clone Cells/cytology , Clone Cells/transplantation , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome
4.
Microcirculation ; 25(5): e12455, 2018 07.
Article in English | MEDLINE | ID: mdl-29665185

ABSTRACT

OBJECTIVE: Endothelial cells form vascular beds in all organs and are exposed to a range of mechanical forces that regulate cellular phenotype. We sought to determine the role of endothelial luminal surface stiffness in tissue-specific mechanotransduction of laminar shear stress in microvascular mouse cells and the role of arachidonic acid in mediating this response. METHODS: Microvascular mouse endothelial cells were subjected to laminar shear stress at 4 dynes/cm2 for 12 hours in parallel plate flow chambers that enabled real-time optical microscopy and atomic force microscopy measurements of cell stiffness. RESULTS: Lung endothelial cells aligned parallel to flow, while cardiac endothelial cells did not. This rapid alignment was accompanied by increased cell stiffness. The addition of arachidonic acid to cardiac endothelial cells increased alignment and stiffness in response to shear stress. Inhibition of arachidonic acid in lung endothelial cells and embryonic stem cell-derived endothelial cells prevented cellular alignment and decreased cell stiffness. CONCLUSIONS: Our findings suggest that increased endothelial luminal surface stiffness in microvascular cells may facilitate mechanotransduction and alignment in response to laminar shear stress. Furthermore, the arachidonic acid pathway may mediate this tissue-specific process. An improved understanding of this response will aid in the treatment of organ-specific vascular disease.


Subject(s)
Endothelial Cells/physiology , Mechanotransduction, Cellular , Stress, Mechanical , Animals , Arachidonic Acid/pharmacology , Biomechanical Phenomena , Cells, Cultured , Lung/cytology , Mice , Microcirculation , Myocardium/cytology , Surface Properties
5.
Radiology ; 285(1): 114-123, 2017 10.
Article in English | MEDLINE | ID: mdl-28498793

ABSTRACT

Purpose To demonstrate the feasibility of imaging-guided catheter-directed delivery of endothelial cell therapy in a porcine model of cirrhosis for liver regeneration. Materials and Methods After approval from the institutional animal care and use committee, autologous liver endothelial cells were grown from core hepatic specimens from swine. Cirrhosis was induced in swine by means of transcatheter infusion of ethanol and iodized oil into the hepatic artery. Three weeks after induction of cirrhosis, the swine were randomly assigned to receive autologous cell therapy (endothelial cells, n = 4) or control treatment (phosphate-buffered saline, n = 4) by means of imaging-guided transhepatic intraportal catheterization. Fluorescence-activated cell sorting analysis was performed on biopsy samples 1 hour after therapy. Three weeks after intraportal delivery of endothelial cells, the swine were euthanized and the explanted liver underwent quantitative pathologic examination. Statistical analysis was performed with an unpaired t test by using unequal variance. Results Liver endothelial cells were successfully isolated, cultured, and expanded from eight 20-mm, 18-gauge hepatic core samples to 50 × 106 autologous cells per pig. Intraportal delivery of endothelial cell therapy or saline was technically successful in all eight swine, with no complications. Endothelial cells were present in the liver for a minimum of 1 hour after intraportal infusion. Swine treated with endothelial cell therapy showed mean levels of surrogate markers of hepatobiliary injury that were consistent with decreases in hepatic fibrosis and biliary ductal damage relative to the control animals, although statistical significance was not met in this pilot study: The mean percentage of positive pixels at Masson trichrome staining was 7.28% vs 5.57%, respectively (P = .20), the mean proliferation index with cytokeratin wide-spectrum was 2.55 vs 1.13 (P = .06), and the mean proliferation index with Ki67 was 7.08 vs 4.96 (P = .14). Conclusion The results confirm the feasibility of imaging-guided catheter-directed endothelial cell therapy with an intraportal technique for the treatment of cirrhosis in a porcine model. A trend toward decreased liver fibrosis with endothelial cell therapy was observed. Larger animal studies and human studies are necessary to confirm significance. © RSNA, 2017.


Subject(s)
Cell Transplantation/methods , Endothelial Cells/transplantation , Liver Cirrhosis/diagnostic imaging , Liver Cirrhosis/therapy , Surgery, Computer-Assisted/methods , Animals , Catheters , Cell- and Tissue-Based Therapy/methods , Disease Models, Animal , Feasibility Studies , Female , Liver/chemistry , Liver/cytology , Liver/diagnostic imaging , Liver Regeneration/physiology , Swine
6.
Blood ; 121(5): 770-80, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23169780

ABSTRACT

UNLABELLED: Several studies have demonstrated that hematopoietic cells originate from endotheliumin early development; however, the phenotypic progression of progenitor cells during human embryonic hemogenesis is not well described. Here, we define the developmental hierarchy among intermediate populations of hematopoietic progenitor cells (HPCs) derived from human embryonic stem cells (hESCs). We genetically modified hESCs to specifically demarcate acquisition of vascular (VE-cadherin) and hematopoietic (CD41a) cell fate and used this dual-reporting transgenic hESC line to observe endothelial to hematopoietic transition by real-time confocal microscopy. Live imaging and clonal analyses revealed a temporal bias in commitment of HPCs that recapitulates discrete waves of lineage differentiation noted during mammalian hemogenesis. Specifically, HPCs isolated at later time points showed reduced capacity to form erythroid/ megakaryocytic cells and exhibited a tendency toward myeloid fate that was enabled by expression of the Notch ligand Dll4 on hESC-derived vascular feeder cells. These data provide a framework for defining HPC lineage potential, elucidate a molecular contribution from the vascular niche in promoting hematopoietic lineage progression, and distinguish unique subpopulations of hemogenic endothelium during hESC differentiation. KEY POINTS: Live imaging of endothelial to hematopoietic conversion identifies distinct subpopulations of hESC-derived hemogenic endothelium. Expression of the Notch ligand DII4 on vascular ECs drives induction of myeloid fate from hESC-derived hematopoietic progenitors.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/metabolism , Endothelial Cells/metabolism , Hematopoietic Stem Cells/metabolism , Transduction, Genetic , Antigens, CD/biosynthesis , Antigens, CD/genetics , Cadherins/biosynthesis , Cadherins/genetics , Coculture Techniques , Embryonic Stem Cells/cytology , Endothelial Cells/cytology , Feeder Cells , Hematopoietic Stem Cells/cytology , Humans , Platelet Membrane Glycoprotein IIb/biosynthesis , Platelet Membrane Glycoprotein IIb/genetics
7.
Stem Cells ; 32(1): 177-90, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23963623

ABSTRACT

The ability to generate and maintain stable in vitro cultures of mouse endothelial cells (ECs) has great potential for genetic dissection of the numerous pathologies involving vascular dysfunction as well as therapeutic applications. However, previous efforts at achieving sustained cultures of primary stable murine vascular cells have fallen short, and the cellular requirements for EC maintenance in vitro remain undefined. In this study, we have generated vascular ECs from mouse embryonic stem (ES) cells and show that active Akt is essential to their survival and propagation as homogeneous monolayers in vitro. These cells harbor the phenotypical, biochemical, and functional characteristics of ECs and expand throughout long-term cultures, while maintaining their angiogenic capacity. Moreover, Akt-transduced embryonic ECs form functional perfused vessels in vivo that anastomose with host blood vessels. We provide evidence for a novel function of Akt in stabilizing EC identity, whereby the activated form of the protein protects mouse ES cell-derived ECs from TGFß-mediated transdifferentiation by downregulating SMAD3. These findings identify a role for Akt in regulating the developmental potential of ES cell-derived ECs and demonstrate that active Akt maintains endothelial identity in embryonic ECs by interfering with active TGFß-mediated processes that would ordinarily usher these cells to alternate fates.


Subject(s)
Embryonic Stem Cells/metabolism , Endothelial Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Embryonic Stem Cells/cytology , Endothelial Cells/cytology , Mice , Signal Transduction , Transforming Growth Factor beta/antagonists & inhibitors
8.
Cells ; 11(19)2022 09 24.
Article in English | MEDLINE | ID: mdl-36230937

ABSTRACT

Alpha-2-macroglobulin (A2M) is a protease inhibitor that regulates extracellular matrix (ECM) stability and turnover. Here, we show that A2M is expressed by endothelial cells (ECs) from human eye choroid. We demonstrate that retinal pigment epithelium (RPE)-conditioned medium induces A2M expression specifically in ECs. Experiments using chemical inhibitors, blocking antibodies, and recombinant proteins revealed a key role of VEGF-A in RPE-mediated A2M induction in ECs. Furthermore, incubation of ECs with RPE-conditioned medium reduces matrix metalloproteinase-2 gelatinase activity of culture supernatants, which is partially restored after A2M knockdown in ECs. We propose that dysfunctional RPE or choroidal blood vessels, as observed in retinal diseases such as age-related macular degeneration, may disrupt the crosstalk mechanism we describe here leading to alterations in the homeostasis of choroidal ECM, Bruch's membrane and visual function.


Subject(s)
Pregnancy-Associated alpha 2-Macroglobulins , Retinal Pigment Epithelium , Antibodies, Blocking , Culture Media, Conditioned , Endothelial Cells , Female , Gelatinases , Humans , Matrix Metalloproteinase 2 , Pregnancy , Protease Inhibitors , Recombinant Proteins , Transcription Factors , Vascular Endothelial Growth Factor A
9.
J Exp Med ; 218(8)2021 08 02.
Article in English | MEDLINE | ID: mdl-34287647

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is marked by airway inflammation and airspace enlargement (emphysema) leading to airflow obstruction and eventual respiratory failure. Microvasculature dysfunction is associated with COPD/emphysema. However, it is not known if abnormal endothelium drives COPD/emphysema pathology and/or if correcting endothelial dysfunction has therapeutic potential. Here, we show the centrality of endothelial cells to the pathogenesis of COPD/emphysema in human tissue and using an elastase-induced murine model of emphysema. Airspace disease showed significant endothelial cell loss, and transcriptional profiling suggested an apoptotic, angiogenic, and inflammatory state. This alveolar destruction was rescued by intravenous delivery of healthy lung endothelial cells. Leucine-rich α-2-glycoprotein-1 (LRG1) was a driver of emphysema, and deletion of Lrg1 from endothelial cells rescued vascular rarefaction and alveolar regression. Hence, targeting endothelial cell biology through regenerative methods and/or inhibition of the LRG1 pathway may represent strategies of immense potential for the treatment of COPD/emphysema.


Subject(s)
Endothelial Cells/pathology , Lung/pathology , Pulmonary Emphysema/pathology , Administration, Intravenous , Animals , Biomarkers/metabolism , Disease Models, Animal , Endothelial Cells/transplantation , Gene Expression Profiling , Gene Expression Regulation , Glycoproteins/metabolism , Humans , Lung/blood supply , Lung/physiopathology , Mice, Inbred C57BL , Neovascularization, Physiologic , Pancreatic Elastase/metabolism , Phenotype , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/physiopathology , Pulmonary Emphysema/genetics , Pulmonary Emphysema/physiopathology , Severity of Illness Index , Smoking , Transcriptome/genetics
10.
Mol Ther Methods Clin Dev ; 20: 703-715, 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33738325

ABSTRACT

Ex vivo hematopoietic stem and progenitor cell (HSPC) expansion platforms are under active development, designed to increase HSPC numbers and thus engraftment ability of allogeneic cord blood grafts or autologous HSPCs for gene therapies. Murine and in vitro models have not correlated well with clinical outcomes of HSPC expansion, emphasizing the need for relevant pre-clinical models. Our rhesus macaque HSPC competitive autologous transplantation model utilizing genetically barcoded HSPC allows direct analysis of the relative short and long-term engraftment ability of lentivirally transduced HSPCs, along with additional critical characteristics such as HSPC clonal diversity and lineage bias. We investigated the impact of ex vivo expansion of macaque HSPCs on the engineered endothelial cell line (E-HUVECs) platform regarding safety, engraftment of transduced and E-HUVEC-expanded HSPC over time compared to non-expanded HSPC for up to 51 months post-transplantation, and both clonal diversity and lineage distribution of output from each engrafted cell source. Short and long-term engraftment were comparable for E-HUVEC expanded and the non-expanded HSPCs in both animals, despite extensive proliferation of CD34+ cells during 8 days of ex vivo culture for the E-HUVEC HSPCs, and optimization of harvesting and infusion of HSPCs co-cultured on E-HUVEC in the second animal. Long-term hematopoietic output from both E-HUVEC expanded and unexpanded HSPCs was highly polyclonal and multilineage. Overall, the comparable HSPC kinetics of macaques to humans, the ability to study post-transplant clonal patterns, and simultaneous multi-arm comparisons of grafts without the complication of interpreting allogeneic effects makes our model ideal to test ex vivo HSPC expansion platforms, particularly for gene therapy applications.

11.
Mol Cell Biol ; 27(18): 6300-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17636030

ABSTRACT

Cooperation between STAT3 and c-Jun in driving transcription during transfection of reporter constructs is well established, and both proteins are present on some interleukin-6 (IL-6) STAT3-dependent promoters on chromosomal loci. We report that small interfering RNA knockdown of c-Jun or c-Fos diminishes IL-6 induction of some but not all STAT3-dependent mRNAs. Specific contact sites in STAT3 responsible for interaction of a domain of STAT3 with c-Jun were known. Here we show that the B-zip domain of c-Jun interacts with STAT3 and that c-Jun mutation R261A or R261D near but not in the DNA binding domain blocks in vitro STAT3-c-Jun interaction and decreases costimulation of transcription in transfection assays. Cooperative binding to DNA of tyrosine-phosphorylated STAT3 and both wild-type and R261A mutant c-Jun was observed. Even c-Jun mutant R261D, which on its own did not bind DNA, bound DNA weakly in the presence of STAT3. We conclude that a functional interaction between STAT3 and c-Jun while bound to chromosomal DNA elements exists and is necessary for driving transcription on at least some STAT3 target genes. Identifying such required interactive protein interfaces should be a stimulus to search for compounds that could ultimately inhibit the activity of STAT3 in tumors dependent on persistently active STAT3.


Subject(s)
Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , STAT3 Transcription Factor/metabolism , Transcription Factor AP-1/metabolism , Amino Acid Sequence , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Glutathione Transferase/metabolism , Humans , Interleukin-6/pharmacology , Liver Neoplasms/pathology , Molecular Sequence Data , Mutation , Plasmids , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-jun/chemistry , Proto-Oncogene Proteins c-jun/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Rats , Receptors, Interleukin-6/metabolism , Recombinant Proteins/metabolism , STAT3 Transcription Factor/chemistry , STAT3 Transcription Factor/genetics , Transcription Factor AP-1/genetics , Transfection
12.
J Exp Med ; 217(6)2020 06 01.
Article in English | MEDLINE | ID: mdl-32196081

ABSTRACT

The activity and survival of retinal photoreceptors depend on support functions performed by the retinal pigment epithelium (RPE) and on oxygen and nutrients delivered by blood vessels in the underlying choroid. By combining single-cell and bulk RNA sequencing, we categorized mouse RPE/choroid cell types and characterized the tissue-specific transcriptomic features of choroidal endothelial cells. We found that choroidal endothelium adjacent to the RPE expresses high levels of Indian Hedgehog and identified its downstream target as stromal GLI1+ mesenchymal stem cell-like cells. In vivo genetic impairment of Hedgehog signaling induced significant loss of choroidal mast cells, as well as an altered inflammatory response and exacerbated visual function defects after retinal damage. Our studies reveal the cellular and molecular landscape of adult RPE/choroid and uncover a Hedgehog-regulated choroidal immunomodulatory signaling circuit. These results open new avenues for the study and treatment of retinal vascular diseases and choroid-related inflammatory blinding disorders.


Subject(s)
Choroid/immunology , Choroid/pathology , Endothelium/immunology , Immunomodulation , Single-Cell Analysis , Animals , Cell Proliferation , Endothelial Cells/metabolism , Gene Expression Regulation , Hedgehog Proteins/metabolism , Inflammation/genetics , Mast Cells/metabolism , Melanocytes/metabolism , Melanocytes/pathology , Mice, Inbred C57BL , Organ Specificity , Retinal Pigment Epithelium/metabolism , Signal Transduction , Transcription, Genetic , Zinc Finger Protein GLI1/metabolism
13.
J Vis Exp ; (135)2018 05 16.
Article in English | MEDLINE | ID: mdl-29863664

ABSTRACT

Infertility is a frequent side effect of chemotherapy and/or radiotherapy and for some patients, cryopreservation of oocytes or embryos is not an option. As an alternative, an increasing number of these patients are choosing to cryopreserve ovarian tissue for autograft following recovery and remission. Despite improvements in outcomes among patients undergoing auto-transplantation of cryopreserved ovarian tissue, efficient revascularization of grafted tissue remains a major obstacle. To mitigate ischemia and thus improve outcomes in patients undergoing auto-transplantation, we developed a vascular cell-based strategy for accelerating perfusion of ovarian tissue. We describe a method for co-transplantation of exogenous endothelial cells (ExECs) with cryopreserved ovarian tissue in a mouse xenograft model. We extend this approach to employ ExECs that have been engineered to constitutively express Anti-Mullerian hormone (AMH), thus enabling sustained paracrine signaling input to ovarian grafts. Co-transplantation with ExECs increased follicular volume and improved antral follicle development, and AMH-expressing ExECs promoted retention of quiescent primordial follicles. This combined strategy may be a useful tool for mitigating ischemia and modulating follicular activation in the context of fertility preservation and/or infertility at large.


Subject(s)
Endothelial Cells/metabolism , Ovary/transplantation , Paracrine Communication/physiology , Tissue Engineering/methods , Animals , Female , Humans , Mice
14.
Sci Immunol ; 3(19)2018 01 12.
Article in English | MEDLINE | ID: mdl-29330161

ABSTRACT

The thymus is not only extremely sensitive to damage but also has a remarkable ability to repair itself. However, the mechanisms underlying this endogenous regeneration remain poorly understood, and this capacity diminishes considerably with age. We show that thymic endothelial cells (ECs) comprise a critical pathway of regeneration via their production of bone morphogenetic protein 4 (BMP4) ECs increased their production of BMP4 after thymic damage, and abrogating BMP4 signaling or production by either pharmacologic or genetic inhibition impaired thymic repair. EC-derived BMP4 acted on thymic epithelial cells (TECs) to increase their expression of Foxn1, a key transcription factor involved in TEC development, maintenance, and regeneration, and its downstream targets such as Dll4, a key mediator of thymocyte development and regeneration. These studies demonstrate the importance of the BMP4 pathway in endogenous tissue regeneration and offer a potential clinical approach to enhance T cell immunity.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Endothelial Cells/metabolism , Regeneration/physiology , Thymus Gland/metabolism , Thymus Gland/physiology , Animals , Cell Proliferation/physiology , Endothelial Cells/physiology , Epithelial Cells/metabolism , Epithelial Cells/physiology , Female , Forkhead Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/physiology , Stem Cells/metabolism , Stem Cells/physiology , T-Lymphocytes/metabolism , T-Lymphocytes/physiology
15.
Nat Commun ; 8: 13963, 2017 01 16.
Article in English | MEDLINE | ID: mdl-28091527

ABSTRACT

Transplanting vascular endothelial cells (ECs) to support metabolism and express regenerative paracrine factors is a strategy to treat vasculopathies and to promote tissue regeneration. However, transplantation strategies have been challenging to develop, because ECs are difficult to culture and little is known about how to direct them to stably integrate into vasculature. Here we show that only amniotic cells could convert to cells that maintain EC gene expression. Even so, these converted cells perform sub-optimally in transplantation studies. Constitutive Akt signalling increases expression of EC morphogenesis genes, including Sox17, shifts the genomic targeting of Fli1 to favour nearby Sox consensus sites and enhances the vascular function of converted cells. Enforced expression of Sox17 increases expression of morphogenesis genes and promotes integration of transplanted converted cells into injured vessels. Thus, Ets transcription factors specify non-vascular, amniotic cells to EC-like cells, whereas Sox17 expression is required to confer EC function.


Subject(s)
Endothelial Cells/transplantation , Endothelium, Vascular/metabolism , SOXF Transcription Factors/metabolism , Vascular Diseases/therapy , Amnion/cytology , Amnion/embryology , Amnion/metabolism , Animals , Endothelial Cells/metabolism , Endothelium, Vascular/physiopathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , Regeneration , SOXF Transcription Factors/genetics , Vascular Diseases/genetics , Vascular Diseases/metabolism , Vascular Diseases/physiopathology
16.
Sci Rep ; 7(1): 8203, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28811567

ABSTRACT

Despite major advances in tissue cryopreservation and auto-transplantation, reperfusion ischemia and hypoxia have been reported as major obstacles to successful recovery of the follicular pool within grafted ovarian tissue. We demonstrate a benefit to follicular survival and function in human ovarian tissue that is co-transplanted with exogenous endothelial cells (ExEC). ExECs were capable of forming functionally perfused vessels at the host/graft interface and increased both viability and follicular volume in ExEC-assisted grafts with resumption of antral follicle development in long-term grafts. ExECs that were engineered to constitutively express anti-mullerian hormone (AMH) induced a greater proportion of quiescent primordial follicles than control ExECs, indicating suppression of premature mobilization that has been noted in the context of ovarian tissue transplantation. These findings present a cell-based strategy that combines accelerated perfusion with direct paracrine delivery of a bioactive payload to transplanted ovarian tissue.


Subject(s)
Endothelial Cells/metabolism , Endothelium/metabolism , Neovascularization, Physiologic , Ovary/physiology , Paracrine Communication , Tissue Engineering , Adolescent , Animals , Biomarkers , Child , Cryopreservation , Female , Fertility Preservation , Fluorescent Antibody Technique , Graft Survival , Humans , Mice , Transplants , Young Adult
17.
Stem Cells Transl Med ; 6(3): 864-876, 2017 03.
Article in English | MEDLINE | ID: mdl-28297579

ABSTRACT

Successful expansion of bone marrow (BM) hematopoietic stem and progenitor cells (HSPCs) would benefit many HSPC transplantation and gene therapy/editing applications. However, current expansion technologies have been limited by a loss of multipotency and self-renewal properties ex vivo. We hypothesized that an ex vivo vascular niche would provide prohematopoietic signals to expand HSPCs while maintaining multipotency and self-renewal. To test this hypothesis, BM autologous CD34+ cells were expanded in endothelial cell (EC) coculture and transplanted in nonhuman primates. CD34+ C38- HSPCs cocultured with ECs expanded up to 17-fold, with a significant increase in hematopoietic colony-forming activity compared with cells cultured with cytokines alone (colony-forming unit-granulocyte-erythroid-macrophage-monocyte; p < .005). BM CD34+ cells that were transduced with green fluorescent protein lentivirus vector and expanded on ECs engrafted long term with multilineage polyclonal reconstitution. Gene marking was observed in granulocytes, lymphocytes, platelets, and erythrocytes. Whole transcriptome analysis indicated that EC coculture altered the expression profile of 75 genes in the BM CD34+ cells without impeding the long-term engraftment potential. These findings show that an ex vivo vascular niche is an effective platform for expansion of adult BM HSPCs. Stem Cells Translational Medicine 2017;6:864-876.


Subject(s)
Bone Marrow Cells/cytology , Endothelial Cells/cytology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Animals , Antigens, CD34/metabolism , Cell Lineage , Cell Proliferation , Endothelial Cells/metabolism , Gene Expression Profiling , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Humans , Primates , Time Factors
18.
Nat Commun ; 8: 15374, 2017 05 19.
Article in English | MEDLINE | ID: mdl-28524846

ABSTRACT

The outer blood-retina barrier is established through the coordinated terminal maturation of the retinal pigment epithelium (RPE), fenestrated choroid endothelial cells (ECs) and Bruch's membrane, a highly organized basement membrane that lies between both cell types. Here we study the contribution of choroid ECs to this process by comparing their gene expression profile before (P5) and after (P30) the critical postnatal period when mice acquire mature visual function. Transcriptome analyses show that expression of extracellular matrix-related genes changes dramatically over this period. Co-culture experiments support the existence of a novel regulatory pathway: ECs secrete factors that remodel RPE basement membrane, and integrin receptors sense these changes triggering Rho GTPase signals that modulate RPE tight junctions and enhance RPE barrier function. We anticipate our results will spawn a search for additional roles of choroid ECs in RPE physiology and disease.


Subject(s)
Basement Membrane/metabolism , Bruch Membrane/metabolism , Extracellular Matrix/metabolism , Retinal Pigment Epithelium/metabolism , Tight Junctions/metabolism , Animals , Biotinylation , Blood-Retinal Barrier/metabolism , Cell Adhesion , Cell Survival , Cells, Cultured , Choroid/metabolism , Coculture Techniques , Electroretinography , Female , Integrins/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Permeability , Protein-Lysine 6-Oxidase/metabolism , RNA, Messenger/metabolism , Sequence Analysis, RNA
19.
Radiat Res ; 186(2): 196-202, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27459700

ABSTRACT

Current therapeutic approaches for treatment of exposure to radiation involve the use of antioxidants, chelating agents, recombinant growth factors and transplantation of stem cells (e.g., hematopoietic stem cell transplantation). However, exposure to high-dose radiation is associated with severe damage to the vasculature of vital organs, often leading to impaired healing, tissue necrosis, thrombosis and defective regeneration caused by aberrant fibrosis. It is very unlikely that infusion of protective chemicals will reverse severe damage to the vascular endothelial cells (ECs). The role of irradiated vasculature in mediating acute and chronic radiation syndromes has not been fully appreciated or well studied. New approaches are necessary to replace and reconstitute ECs in organs that are irreversibly damaged by radiation. We have set forth the novel concept that ECs provide paracrine signals, also known as angiocrine signals, which not only promote healing of irradiated tissue but also direct organ regeneration without provoking fibrosis. We have developed innovative technologies that enable manufacturing and banking of human GMP-grade ECs. These ECs can be transplanted intravenously to home to and engraft to injured tissues where they augment organ repair, while preventing maladaptive fibrosis. In the past, therapeutic transplantation of ECs was not possible due to a shortage of availability of suitable donor cell sources and preclinical models, a lack of understanding of the immune privilege of ECs, and inadequate methodologies for expansion and banking of engraftable ECs. Recent advances made by our group as well as other laboratories have breached the most significant of these obstacles with the development of technologies to manufacture clinical-scale quantities of GMP-grade and human ECs in culture, including genetically diverse reprogrammed human amniotic cells into vascular ECs (rAC-VECs) or human pluripotent stem cells into vascular ECs (iVECs). This approach provides a path to therapeutic EC transplantation that can be infused concomitantly or sequentially with hematopoietic stem cell transplantation more than 24 h after irradiation to support multi-organ regeneration, thereby improving immediate and long-term survival, while limiting long-term morbidity resulting from nonregenerative damage repair pathways.


Subject(s)
Cell Transplantation/methods , Endothelial Cells/transplantation , Radiation Injuries/pathology , Radiation Injuries/therapy , Acute Disease , Animals , Chronic Disease , Humans
20.
Nat Med ; 22(2): 154-62, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26779814

ABSTRACT

Although the lung can undergo self-repair after injury, fibrosis in chronically injured or diseased lungs can occur at the expense of regeneration. Here we study how a hematopoietic-vascular niche regulates alveolar repair and lung fibrosis. Using intratracheal injection of bleomycin or hydrochloric acid in mice, we show that repetitive lung injury activates pulmonary capillary endothelial cells (PCECs) and perivascular macrophages, impeding alveolar repair and promoting fibrosis. Whereas the chemokine receptor CXCR7, expressed on PCECs, acts to prevent epithelial damage and ameliorate fibrosis after a single round of treatment with bleomycin or hydrochloric acid, repeated injury leads to suppression of CXCR7 expression and recruitment of vascular endothelial growth factor receptor 1 (VEGFR1)-expressing perivascular macrophages. This recruitment stimulates Wnt/ß-catenin-dependent persistent upregulation of the Notch ligand Jagged1 (encoded by Jag1) in PCECs, which in turn stimulates exuberant Notch signaling in perivascular fibroblasts and enhances fibrosis. Administration of a CXCR7 agonist or PCEC-targeted Jag1 shRNA after lung injury promotes alveolar repair and reduces fibrosis. Thus, targeting of a maladapted hematopoietic-vascular niche, in which macrophages, PCECs and perivascular fibroblasts interact, may help to develop therapy to spur lung regeneration and alleviate fibrosis.


Subject(s)
Calcium-Binding Proteins/metabolism , Capillaries/metabolism , Endothelial Cells/metabolism , Fibroblasts/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lung Injury/metabolism , Lung/metabolism , Membrane Proteins/metabolism , Pulmonary Fibrosis/metabolism , Receptors, CXCR/metabolism , Regeneration/physiology , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Calcium-Binding Proteins/antagonists & inhibitors , Capillaries/drug effects , Endothelial Cells/drug effects , Endothelial Cells/physiology , Fibroblasts/drug effects , Fibrosis , Fluorescent Antibody Technique , Humans , Hydrochloric Acid/toxicity , Jagged-1 Protein , Lung/drug effects , Lung/pathology , Lung/physiology , Macrophages/drug effects , Macrophages/metabolism , Membrane Proteins/antagonists & inhibitors , Mice , Oligopeptides/pharmacology , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pulmonary Circulation/drug effects , Pulmonary Circulation/physiology , RNA, Small Interfering/pharmacology , Receptors, CXCR/agonists , Receptors, Notch/metabolism , Regeneration/drug effects , Serrate-Jagged Proteins , Smad3 Protein/drug effects , Smad3 Protein/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Wnt Signaling Pathway
SELECTION OF CITATIONS
SEARCH DETAIL