Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Biochem Biophys Res Commun ; 518(2): 227-232, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31416613

ABSTRACT

INTRODUCTION: Receptor tyrosine kinases have been implicated in various vascular remodeling processes and cardiovascular disease. However, their role in the regulation of vascular tone is poorly understood. Herein, we evaluate the contribution of c-Kit signaling to vasoactive responses. METHODS: The vascular reactivity of mesenteric arteries was assessed under isobaric conditions in c-Kit deficient (KitW/W-v) and littermate control mice (Kit+/+) using pressure myography. Protein levels of soluble guanylyl cyclase beta 1 (sGCß1) were quantified by Western blot. Mean arterial pressure was measured after high salt (8% NaCl) diet treatment using the tail-cuff method. RESULTS: Smooth muscle cells (SMCs) from c-Kit deficient mice showed a 5-fold downregulation of sGCß1 compared to controls. Endothelium-dependent relaxation of mesenteric arteries demonstrated a predominance of prostanoid vs. nitric oxide (NO) signaling in both animal groups. The dependence on prostanoid-induced dilation was higher in c-Kit mutant mice than in controls, as indicated by a significant impairment in vasorelaxation with indomethacin with respect to the latter. Endothelium-independent relaxation showed significant dysfunction of NO signaling in c-Kit deficient SMCs compared to controls. Mesenteric artery dilation was rescued by addition of a cGMP analog, but not with a NO donor, indicating a deficiency in cGMP production in c-Kit deficient SMCs. Finally, c-Kit deficient mice developed higher blood pressure on an 8% NaCl diet compared to their control littermates. CONCLUSION: c-Kit deficiency inhibits NO signaling in SMCs. The existence of this c-Kit/sGC signaling axis may be relevant for vascular reactivity and remodeling.


Subject(s)
Myocytes, Smooth Muscle/metabolism , Nitric Oxide/metabolism , Proto-Oncogene Proteins c-kit/deficiency , Signal Transduction , Animals , Arteries/drug effects , Arteries/physiology , Blood Pressure/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Mice , Prostaglandins/pharmacology , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction/drug effects , Sodium Chloride, Dietary , Vasodilation/drug effects
2.
Cell Mol Life Sci ; 76(2): 369-380, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30357422

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) represent the lifelong source of all blood cells and continuously regenerate the hematopoietic system through differentiation and self-renewal. The process of differentiation is initiated in the G1 phase of the cell cycle, when stem cells leave their quiescent state. During G1, the anaphase-promoting complex or cyclosome associated with the coactivator Cdh1 is highly active and marks proteins for proteasomal degradation to regulate cell proliferation. Following Cdh1 knockdown in HSPCs, we analyzed human and mouse hematopoiesis in vitro and in vivo in competitive transplantation assays. We found that Cdh1 is highly expressed in human CD34+ HSPCs and downregulated in differentiated subsets; whereas, loss of Cdh1 restricts myeloid differentiation, supports B cell development and preserves immature short-term HSPCs without affecting proliferation or viability. Our data highlight a role of Cdh1 as a regulator of balancing the maintenance of HSPCs and differentiation into mature blood cells.


Subject(s)
Cdh1 Proteins/metabolism , Cell Differentiation/genetics , Hematopoietic Stem Cells/cytology , Animals , Antigens, CD34/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cdh1 Proteins/antagonists & inhibitors , Cdh1 Proteins/genetics , Cell Proliferation , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , RNA Interference , RNA, Small Interfering/metabolism , Stem Cells/cytology , Stem Cells/metabolism
3.
Blood ; 131(21): e1-e11, 2018 05 24.
Article in English | MEDLINE | ID: mdl-29588278

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) maintain the adult blood system, and their dysregulation causes a multitude of diseases. However, the differentiation journeys toward specific hematopoietic lineages remain ill defined, and system-wide disease interpretation remains challenging. Here, we have profiled 44 802 mouse bone marrow HSPCs using single-cell RNA sequencing to provide a comprehensive transcriptional landscape with entry points to 8 different blood lineages (lymphoid, megakaryocyte, erythroid, neutrophil, monocyte, eosinophil, mast cell, and basophil progenitors). We identified a common basophil/mast cell bone marrow progenitor and characterized its molecular profile at the single-cell level. Transcriptional profiling of 13 815 HSPCs from the c-Kit mutant (W41/W41) mouse model revealed the absence of a distinct mast cell lineage entry point, together with global shifts in cell type abundance. Proliferative defects were accompanied by reduced Myc expression. Potential compensatory processes included upregulation of the integrated stress response pathway and downregulation of proapoptotic gene expression in erythroid progenitors, thus providing a template of how large-scale single-cell transcriptomic studies can bridge between molecular phenotypes and quantitative population changes.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mutation , Proto-Oncogene Proteins c-kit/deficiency , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Line, Tumor , Cells, Cultured , Gene Expression Profiling , Mice , Mice, Knockout , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction , Single-Cell Analysis , Transcriptome
4.
Surgery ; 163(4): 877-882, 2018 04.
Article in English | MEDLINE | ID: mdl-29287914

ABSTRACT

BACKGROUND: Arteriogenesis is a process whereby collateral vessels remodel usually in response to increased blood flow and/or wall stress. Remodeling of collaterals can function as a natural bypass to alleviate ischemia during arterial occlusion. Here we used a genetic approach to investigate possible roles of tyrosine receptor c-Kit in arteriogenesis. METHODS: Mutant mice with loss of c-Kit function (KitW/W-v), and controls were subjected to hindlimb ischemia. Blood flow recovery was evaluated pre-, post-, and weekly after ischemia. Foot ischemic damage and function were assessed between days 1 to 14 post-ischemia while collaterals remodeling were measured 28 days post-ischemia. Both groups of mice also were subjected to wild type bone marrow cells transplantation 3 weeks before hindlimb ischemia to evaluate possible contributions of defective bone marrow c-Kit expression on vascular recovery. RESULTS: KitW/W-v mice displayed impaired blood flow recovery, greater ischemic damage and foot dysfunction after ischemia compared to controls. KitW/W-v mice also demonstrated impaired collateral remodeling consistent with flow recovery findings. Because arteriogenesis is a biological process that involves bone marrow-derived cells, we investigated which source of c-Kit signaling (bone marrow or vascular) plays a major role in arteriogenesis. KitW/W-v mice transplanted with bone marrow wild type cells exhibited similar phenotype of impaired blood flow recovery, greater tissue ischemic damage and foot dysfunction as nontransplanted KitW/W-v mice. CONCLUSION: This study provides evidence that c-Kit signaling is required during arteriogenesis. Also, it strongly suggests a vascular role for c-Kit signaling because rescue of systemic c-Kit activity by bone marrow transplantation did not augment the functional recovery of KitW/W-v mouse hindlimbs.


Subject(s)
Collateral Circulation/physiology , Hindlimb/blood supply , Ischemia/physiopathology , Neovascularization, Physiologic/physiology , Proto-Oncogene Proteins c-kit/deficiency , Animals , Biomarkers/metabolism , Bone Marrow Transplantation , Hindlimb/physiology , Hindlimb/physiopathology , Ischemia/metabolism , Ischemia/therapy , Male , Mice , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction
5.
Stem Cell Reports ; 9(4): 1024-1033, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28943250

ABSTRACT

Generation of hematopoietic stem cells (HSCs) from pluripotent stem cells (PSCs) could potentially provide unlimited HSCs for clinical transplantation, a curative treatment for numerous blood diseases. However, to date, bona fide HSC generation has been largely unsuccessful in vitro. We have previously described proof of concept for in vivo HSC generation from PSCs via teratoma formation. However, our first-generation system was complex and the output low. Here, we further optimize this technology and demonstrate the following: (1) simplified HSC generation using transcription factor overexpression; (2) improved HSC output using c-Kit-deficient host mice, and (3) that teratomas can be transplanted and cryopreserved. We demonstrate that overexpression of Gfi1b, c-Fos, and Gata2, previously reported to transdifferentiate fibroblasts into hematopoietic progenitors in vitro, can induce long-term HSC formation in vivo. Our in vivo system provides a useful platform to investigate new strategies and re-evaluate existing strategies to generate HSCs and study HSC development.


Subject(s)
Cell Transdifferentiation/genetics , GATA2 Transcription Factor/genetics , Gene Expression , Genes, fos , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Teratoma/genetics , Animals , Biomarkers , Cellular Reprogramming/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Order , Genetic Vectors/genetics , Immunophenotyping , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Mice , Proto-Oncogene Proteins c-kit/deficiency , Stem Cell Transplantation , Teratoma/metabolism , Teratoma/pathology
6.
Circ Res ; 121(8): 941-950, 2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28739590

ABSTRACT

RATIONALE: Deep vein thrombosis (DVT) and its complication pulmonary embolism have high morbidity reducing quality of life and leading to death. Cellular mechanisms of DVT initiation remain poorly understood. OBJECTIVE: We sought to determine the role of mast cells (MCs) in DVT initiation and validate MCs as a potential target for DVT prevention. METHODS AND RESULTS: In a mouse model, DVT was induced by partial ligation (stenosis) of the inferior vena cava. We demonstrated that 2 strains of mice deficient for MCs were completely protected from DVT. Adoptive transfer of in vitro differentiated MCs restored thrombosis. MCs were present in the venous wall, and the number of granule-containing MCs decreased with thrombosis. Pharmacological depletion of MCs granules or prevention of MC degranulation also reduced DVT. Basal plasma levels of von Willebrand factor and recruitment of platelets to the inferior vena cava wall after DVT induction were reduced in MC-deficient mice. Stenosis application increased plasma levels of soluble P-selectin in wild-type but not in MC-deficient mice. MC releasate elevated ICAM-1 (intercellular adhesion molecule-1) expression on HUVEC (human umbilical vein endothelial cells) in vitro. Topical application of compound 48/80, an MC secretagogue, or histamine, a Weibel-Palade body secretagogue from MCs, potentiated DVT in wild-type mice, and histamine restored thrombosis in MC-deficient animals. CONCLUSIONS: MCs exacerbate DVT likely through endothelial activation and Weibel-Palade body release, which is, at least in part, mediated by histamine. Because MCs do not directly contribute to normal hemostasis, they can be considered potential targets for prevention of DVT in humans.


Subject(s)
Blood Coagulation , Cell Degranulation , Histamine/metabolism , Mast Cells/metabolism , Vena Cava, Inferior/metabolism , Venous Thrombosis/metabolism , Adoptive Transfer , Animals , Blood Coagulation/drug effects , Blood Platelets/metabolism , Cell Degranulation/drug effects , Cells, Cultured , Disease Models, Animal , Fibrinolytic Agents/pharmacology , Genetic Predisposition to Disease , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Ligation , Mast Cells/drug effects , Mast Cells/transplantation , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , Selenoprotein P/metabolism , Signal Transduction , Vena Cava, Inferior/drug effects , Vena Cava, Inferior/surgery , Venous Thrombosis/blood , Venous Thrombosis/genetics , Venous Thrombosis/prevention & control , Weibel-Palade Bodies/metabolism , von Willebrand Factor/metabolism
7.
Blood ; 129(17): 2384-2394, 2017 04 27.
Article in English | MEDLINE | ID: mdl-28122740

ABSTRACT

Understanding leukemia heterogeneity is critical for the development of curative treatments as the failure to eliminate therapy-persistent leukemic stem cells (LSCs) may result in disease relapse. Here we have combined high-throughput immunophenotypic screens with large-scale single-cell gene expression analysis to define the heterogeneity within the LSC population in chronic phase chronic myeloid leukemia (CML) patients at diagnosis and following conventional tyrosine kinase inhibitor (TKI) treatment. Our results reveal substantial heterogeneity within the putative LSC population in CML at diagnosis and demonstrate differences in response to subsequent TKI treatment between distinct subpopulations. Importantly, LSC subpopulations with myeloid and proliferative molecular signatures are proportionally reduced at a higher extent in response to TKI therapy compared with subfractions displaying primitive and quiescent signatures. Additionally, cell surface expression of the CML stem cell markers CD25, CD26, and IL1RAP is high in all subpopulations at diagnosis but downregulated and unevenly distributed across subpopulations in response to TKI treatment. The most TKI-insensitive cells of the LSC compartment can be captured within the CD45RA- fraction and further defined as positive for CD26 in combination with an aberrant lack of cKIT expression. Together, our results expose a considerable heterogeneity of the CML stem cell population and propose a Lin-CD34+CD38-/lowCD45RA-cKIT-CD26+ population as a potential therapeutic target for improved therapy response.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Neoplastic Stem Cells/drug effects , Protein Kinase Inhibitors/therapeutic use , Single-Cell Analysis/methods , ADP-ribosyl Cyclase 1/deficiency , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/immunology , Antigens, CD34/genetics , Antigens, CD34/immunology , Biomarkers, Tumor/immunology , Case-Control Studies , Cell Lineage/immunology , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/immunology , Gene Expression , Genetic Heterogeneity , Humans , Immunophenotyping , Interleukin-1 Receptor Accessory Protein/genetics , Interleukin-1 Receptor Accessory Protein/immunology , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukocyte Common Antigens/deficiency , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/immunology , Treatment Outcome
8.
PLoS One ; 11(12): e0168772, 2016.
Article in English | MEDLINE | ID: mdl-28002455

ABSTRACT

Bladder inflammation frequently causes cystitis pain and lower urinary tract dysfunction (LUTD) such as urinary frequency and urgency. Although mast cells have been identified to play a critical role in bladder inflammation and pain, the role of mast cells in cystitis-associated LUTD has not been demonstrated. Interstitial cystitis/bladder pain syndrome (IC/BPS) is a chronic and debilitating inflammatory condition of the urinary bladder characterized by the hallmark symptoms of pelvic pain and LUTD. In this study we investigated the role of mast cells in LUTD using a transgenic autoimmune cystitis model (URO-OVA) that reproduces many clinical correlates of IC/BPS. URO-OVA mice express the membrane form of the model antigen ovalbumin (OVA) as a self-antigen on the urothelium and develop bladder inflammation upon introduction of OVA-specific T cells. To investigate the role of mast cells, we crossed URO-OVA mice with mast cell-deficient KitW-sh mice to generate URO-OVA/KitW-sh mice that retained urothelial OVA expression but lacked endogenous mast cells. We compared URO-OVA mice with URO-OVA/KitW-sh mice with and without mast cell reconstitution in response to cystitis induction. URO-OVA mice developed profound bladder inflammation with increased mast cell counts and LUTD, including increased total number of voids, decreased mean volume voided per micturition, and decreased maximum volume voided per micturition, after cystitis induction. In contrast, similarly cystitis-induced URO-OVA/KitW-sh mice developed reduced bladder inflammation with no mast cells and LUTD detected. However, after mast cell reconstitution URO-OVA/KitW-sh mice restored the ability to develop bladder inflammation and LUTD following cystitis induction. We further treated URO-OVA mice with cromolyn, a mast cell membrane stabilizer, and found that cromolyn treatment reversed bladder inflammation and LUTD in the animal model. Our results provide direct evidence for the role of mast cells in cystitis-associated LUTD, supporting the use of mast cell inhibitors for treatment of certain forms of IC/BPS.


Subject(s)
Cystitis, Interstitial/etiology , Mast Cells/metabolism , Pelvic Pain/etiology , Animals , Autoimmune Diseases/etiology , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Behavior, Animal/physiology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cromolyn Sodium/pharmacology , Cystitis, Interstitial/immunology , Cystitis, Interstitial/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Mast Cells/cytology , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/immunology , Pelvic Pain/immunology , Pelvic Pain/metabolism , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , RNA, Messenger/metabolism , Urinary Bladder/drug effects , Urinary Bladder/metabolism , Urinary Bladder/pathology
9.
Stem Cell Reports ; 7(4): 591-601, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27618723

ABSTRACT

Human erythro-megakaryopoiesis does not occur in humanized mouse models, preventing the in vivo analysis of human hematopoietic stem cell (HSC) differentiation into these lineages in a surrogate host. Here we show that stably engrafted KIT-deficient NOD/SCID Il2rg-/-KitW41/W41 (NSGW41) mice support much improved human erythropoiesis and platelet formation compared with irradiated NSG recipients. Considerable numbers of human erythroblasts and mature thrombocytes are present in the bone marrow and blood, respectively. Morphology, composition, and enucleation capacity of de novo generated human erythroblasts in NSGW41 mice are comparable with those in human bone marrow. Overexpression of human erythropoietin showed no further improvement in human erythrocyte output, but depletion of macrophages led to the appearance of human erythrocytes in the blood. Human erythropoiesis up to normoblasts and platelet formation is fully supported in NSGW41 mice, allowing the analysis of human HSC differentiation into these lineages, the exploration of certain pathophysiologies, and the evaluation of gene therapeutic approaches.


Subject(s)
Erythropoiesis/genetics , Thrombopoiesis/genetics , Animals , Cell Differentiation/genetics , Cord Blood Stem Cell Transplantation , Erythroblasts/cytology , Erythroblasts/metabolism , Erythropoietin/genetics , Erythropoietin/pharmacology , Gene Expression , Graft Survival , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Heterografts , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics
10.
Sci Rep ; 6: 32100, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27572504

ABSTRACT

Adult neurogenesis rarely occurs in the enteric nervous system (ENS). In this study, we demonstrated that, after intestinal myenteric plexus (MP) ablation with benzalkonium chloride (BAC), adult neurogenesis in the ENS was significantly induced in c-kit loss-of-function mutant mice (W/W(v)). Almost all neurons and fibers in the MP disappeared after BAC treatment. However, 1 week after ablation, substantial penetration of nerve fibers from the non-damaged area was observed in the MP, longitudinal muscle and subserosal layers in both wildtype and W/W(v) mice. Two weeks after BAC treatment, in addition to the penetrating fibers, a substantial number of ectopic neurons appeared in the subserosal and longitudinal muscle layers of W/W(v) mice, whereas only a few ectopic neurons appeared in wildtype mice. Such ectopic neurons expressed either excitatory or inhibitory intrinsic motor neuron markers and formed ganglion-like structures, including glial cells, synaptic vesicles and basal lamina. Furthermore, oral administration of imatinib, an inhibitor of c-Kit and an anticancer agent for gastrointestinal stromal tumors, markedly induced appearance of ectopic neurons after BAC treatment, even in wildtype mice. These results suggest that adult neurogenesis in the ENS is negatively regulated by c-Kit signaling in vivo.


Subject(s)
Enteric Nervous System/metabolism , Loss of Function Mutation , Neurogenesis/physiology , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Animals , Benzalkonium Compounds/pharmacology , Enteric Nervous System/drug effects , Enteric Nervous System/pathology , Ileum/innervation , Imatinib Mesylate/pharmacology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Muscle, Smooth/pathology , NADP/analysis , Nerve Fibers/pathology , Neurogenesis/drug effects , Neurogenesis/genetics , Neurons/chemistry , Neurons/pathology , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , Signal Transduction/drug effects , Signal Transduction/physiology
11.
Eur J Haematol ; 97(3): 261-70, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26660446

ABSTRACT

Heterozygous mutations in the transcriptional regulator GATA-2 associate with multilineage immunodeficiency, myelodysplastic syndrome (MDS), and acute myeloid leukemia (AML). The majority of these mutations localize in the zinc finger (ZnF) domains, which mediate GATA-2 DNA binding. Deregulated hematopoiesis with GATA-2 mutation frequently develops in adulthood, yet GATA-2 function in the bone marrow remains unresolved. To investigate this, we conditionally deleted the GATA-2 C-terminal ZnF (C-ZnF) coding sequences in adult mice. Upon Gata2 C-ZnF deletion, we observed rapid peripheral cytopenia, bone marrow failure, and decreased c-Kit expression on hematopoietic progenitors. Transplant studies indicated GATA-2 has a cell-autonomous role in bone marrow hematopoiesis. Moreover, myeloid lineage populations were particularly sensitive to Gata2 hemizygosity, while molecular assays indicated GATA-2 regulates c-Kit expression in multilineage progenitor cells. Enforced c-Kit expression in Gata2 C-ZnF-deficient hematopoietic progenitors enhanced myeloid colony activity, suggesting GATA-2 sustains myelopoiesis via a cell intrinsic role involving maintenance of c-Kit expression. Our results provide insight into mechanisms regulating hematopoiesis in bone marrow and may contribute to a better understanding of immunodeficiency and bone marrow failure associated with GATA-2 mutation.


Subject(s)
Anemia, Aplastic/genetics , Bone Marrow Diseases/genetics , Bone Marrow/pathology , GATA2 Transcription Factor/genetics , Hemoglobinuria, Paroxysmal/genetics , Protein Interaction Domains and Motifs/genetics , Proto-Oncogene Proteins c-kit/deficiency , Sequence Deletion , Zinc Fingers/genetics , Anemia, Aplastic/diagnosis , Anemia, Aplastic/metabolism , Anemia, Aplastic/mortality , Animals , Biomarkers , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Marrow Diseases/diagnosis , Bone Marrow Diseases/metabolism , Bone Marrow Diseases/mortality , Bone Marrow Failure Disorders , Bone and Bones/pathology , Chromatin Immunoprecipitation , Decalcification, Pathologic/genetics , Disease Models, Animal , GATA2 Transcription Factor/chemistry , GATA2 Transcription Factor/metabolism , Gene Expression , Gene Expression Regulation , Gene Frequency , Genes, Reporter , Genotype , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Hemoglobinuria, Paroxysmal/diagnosis , Hemoglobinuria, Paroxysmal/metabolism , Hemoglobinuria, Paroxysmal/mortality , High-Throughput Nucleotide Sequencing , Immunophenotyping , Mice , Mice, Knockout , Prognosis , Side-Population Cells
12.
Stem Cells Transl Med ; 5(2): 164-74, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26683871

ABSTRACT

Several progenitor cell populations have been reported to exist in hearts that play a role in cardiac turnover and/or repair. Despite the presence of cardiac stem and progenitor cells within the myocardium, functional repair of the heart after injury is inadequate. Identification of the signaling pathways involved in the expansion and differentiation of cardiac progenitor cells (CPCs) will broaden insight into the fundamental mechanisms playing a role in cardiac homeostasis and disease and might provide strategies for in vivo regenerative therapies. To understand and exploit cardiac ontogeny for drug discovery efforts, we developed an in vitro human induced pluripotent stem cell-derived CPC model system using a highly enriched population of KDR(pos)/CKIT(neg)/NKX2.5(pos) CPCs. Using this model system, these CPCs were capable of generating highly enriched cultures of cardiomyocytes under directed differentiation conditions. In order to facilitate the identification of pathways and targets involved in proliferation and differentiation of resident CPCs, we developed phenotypic screening assays. Screening paradigms for therapeutic applications require a robust, scalable, and consistent methodology. In the present study, we have demonstrated the suitability of these cells for medium to high-throughput screens to assess both proliferation and multilineage differentiation. Using this CPC model system and a small directed compound set, we identified activin-like kinase 5 (transforming growth factor-ß type 1 receptor kinase) inhibitors as novel and potent inducers of human CPC differentiation to cardiomyocytes. Significance: Cardiac disease is a leading cause of morbidity and mortality, with no treatment available that can result in functional repair. This study demonstrates how differentiation of induced pluripotent stem cells can be used to identify and isolate cell populations of interest that can translate to the adult human heart. Two separate examples of phenotypic screens are discussed, demonstrating the value of this biologically relevant and reproducible technology. In addition, this assay system was able to identify novel and potent inducers of differentiation and proliferation of induced pluripotent stem cell-derived cardiac progenitor cells.


Subject(s)
Cell Differentiation/drug effects , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/pharmacology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Biomarkers/metabolism , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Gene Expression , High-Throughput Screening Assays , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Phenotype , Plasmids/chemistry , Plasmids/metabolism , Primary Cell Culture , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Small Molecule Libraries/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
13.
Cancer Res ; 75(18): 3760-70, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26206557

ABSTRACT

Mast cells (MC) are immune cells located next to the intestinal epithelium with regulatory function in maintaining the homeostasis of the mucosal barrier. We have investigated MC activities in colon inflammation and cancer in mice either wild-type (WT) or MC-deficient (Kit(W-sh)) reconstituted or not with bone marrow-derived MCs. Colitis was chemically induced with dextran sodium sulfate (DSS). Tumors were induced by administering azoxymethane (AOM) intraperitoneally before DSS. Following DSS withdrawal, Kit(W-sh) mice showed reduced weight gain and impaired tissue repair compared with their WT littermates or Kit(W-sh) mice reconstituted with bone marrow-derived MCs. MCs were localized in areas of mucosal healing rather than damaged areas where they degraded IL33, an alarmin released by epithelial cells during tissue damage. Kit(W-sh) mice reconstituted with MC deficient for mouse mast cell protease 4 did not restore normal mucosal healing or reduce efficiently inflammation after DSS withdrawal. In contrast with MCs recruited during inflammation-associated wound healing, MCs adjacent to transformed epithelial cells acquired a protumorigenic profile. In AOM- and DSS-treated WT mice, high MC density correlated with high-grade carcinomas. In similarly treated Kit(W-sh) mice, tumors were less extended and displayed lower histologic grade. Our results indicate that the interaction of MCs with epithelial cells is dependent on the inflammatory stage, and on the activation of the tissue repair program. Selective targeting of MCs for prevention or treatment of inflammation-associated colon cancer should be timely pondered to allow tissue repair at premalignant stages or to reduce aggressiveness at the tumor stage.


Subject(s)
Carcinoma/immunology , Colitis/immunology , Colonic Neoplasms/immunology , Intestinal Mucosa/physiology , Mast Cells/physiology , Regeneration/immunology , Animals , Animals, Congenic , Azoxymethane/toxicity , Carcinoma/chemically induced , Carcinoma/pathology , Cell Count , Cell Transformation, Neoplastic/immunology , Cells, Cultured , Colitis/chemically induced , Colitis/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Dextran Sulfate/toxicity , Epithelial Cells/pathology , Humans , Inflammatory Bowel Diseases/pathology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33/physiology , Mast Cells/transplantation , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , Receptors, Interleukin/physiology , Serine Endopeptidases/deficiency , Species Specificity , Specific Pathogen-Free Organisms
14.
Exp Hematol ; 43(7): 578-85, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25892186

ABSTRACT

Identification and isolation of hematopoietic stem cells (HSCs) in mice is most commonly based on the expression of surface molecules Kit and Sca-1 and the absence of markers of mature lineages. However, Sca-1 is absent or weakly expressed in hematopoietic progenitors in many strains, including nonobese diabetic (NOD), BALB/c, C3H, and CBA mice. In addition, both Kit and Sca-1 levels are modulated following bone marrow injury. In these cases, other markers and dye exclusion methods have been employed to identify HSCs, yet there is no antibody-based stain that enables identification of HSCs and early progenitors when Kit and Sca-1 are inadequate. CD201 is a marker that is highly restricted to HSCs and progenitors, and CD27 is expressed at moderate-to-high levels on HSCs. We show here that combining CD201 and CD27 enables highly efficient isolation of long-term HSCs in NOD mice as well as in other strains, including SJL, FVB, AKR, BALB/c, C3H, and CBA. We also find that HSCs appear to maintain expression of CD201 and CD27 after hematopoietic injury when Kit expression is downregulated. These results suggest a widely applicable yet simple alternative for HSC isolation in settings where Kit and Sca-1 expression are insufficient.


Subject(s)
Blood Cells/chemistry , Cell Separation/methods , Flow Cytometry/methods , Hematopoietic Stem Cells/chemistry , Mice, Inbred Strains/blood , Receptors, Cell Surface/blood , Tumor Necrosis Factor Receptor Superfamily, Member 7/blood , Animals , Antigens, Ly/genetics , Antigens, Ly/physiology , Autoimmunity , Blood Cells/cytology , Bone Marrow/radiation effects , Cell Lineage , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Endothelial Protein C Receptor , Gene Expression , Hematopoietic Stem Cells/cytology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred NOD , Mice, Inbred Strains/genetics , Mice, Transgenic , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/physiology , Radiation Chimera , Radiation Injuries, Experimental/blood , Radiation Injuries, Experimental/pathology , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/biosynthesis , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics
15.
World J Gastroenterol ; 20(27): 9106-15, 2014 Jul 21.
Article in English | MEDLINE | ID: mdl-25083083

ABSTRACT

AIM: To test the role of mast cells in gut inflammation and colitis using interleukin (IL)-10-deficient mice as an experimental model. METHODS: Mast cell-deficient (Kit (W-sh/W-sh) ) mice were crossbred with IL-10-deficient mice to obtain double knockout (DKO) mice. The growth, mucosal damage and colitis status of DKO mice were compared with their IL-10-deficient littermates. RESULTS: DKO mice exhibited exacerbated colitis compared with their IL-10-deficient littermates, as shown by increased pathological score, higher myeloperoxidase content, enhanced Th1 type pro-inflammatory cytokines and inflammatory signaling, elevated oxidative stress, as well as pronounced goblet cell loss. In addition, deficiency in mast cells resulted in enhanced mucosal damage, increased gut permeability, and impaired epithelial tight junctions. Mast cell deficiency was also linked to systemic inflammation, as demonstrated by higher serum levels of tumor necrosis factor α and interferon γ in DKO mice than that in IL-10-deficient mice. CONCLUSION: Mast cell deficiency in IL-10-deficient mice resulted in systematic and gut inflammation, impaired gut barrier function, and severer Th1-mediated colitis when compared to mice with only IL-10-deficiency. Inflammation and impaired gut epithelial barrier function likely form a vicious cycle to worsen colitis in the DKO mice.


Subject(s)
Colitis/metabolism , Colon/metabolism , Inflammatory Bowel Diseases/metabolism , Interleukin-10/deficiency , Intestinal Mucosa/metabolism , Mast Cells/metabolism , Animals , Colitis/genetics , Colitis/immunology , Colitis/microbiology , Colitis/pathology , Colon/immunology , Colon/pathology , Disease Models, Animal , Feces/microbiology , Genotype , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Interferon-gamma/metabolism , Interleukin-10/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Male , Mast Cells/immunology , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Peroxidase/metabolism , Phenotype , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , Signal Transduction , Th1 Cells/immunology , Th1 Cells/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism
16.
PLoS One ; 9(1): e84417, 2014.
Article in English | MEDLINE | ID: mdl-24489649

ABSTRACT

Malignant melanoma is a life-threatening skin cancer increasingly diagnosed in the western world. In advanced disease the prognosis is grave. Growth and metastasis formation in melanomas are regulated by a network of cytokines, cytokine-receptors, and adhesion molecules. However, little is known about surface antigens and target expression profiles in human melanomas. We examined the cell surface antigen profile of human skin melanoma cells by multicolor flow cytometry, and compared their phenotype with 4 melanoma cell lines (A375, 607B, Mel-Juso, SK-Mel28). Melanoma cells were defined as CD45-/CD31- cells co-expressing one or more melanoma-related antigens (CD63, CD146, CD166). In most patients, melanoma cells exhibited ErbB3/Her3, CD44/Pgp-1, ICAM-1/CD54 and IGF-1-R/CD221, but did not express CD20, ErbB2/Her2, KIT/CD117, AC133/CD133 or MDR-1/CD243. Melanoma cell lines were found to display a similar phenotype. In most patients, a distinct subpopulation of melanoma cells (4-40%) expressed the erythropoietin receptor (EPO-R) and ErbB4 together with PD-1 and NGF-R/CD271. Both the EPO-R+ and EPO-R- subpopulations produced melanoma lesions in NOD/SCID IL-2Rgamma(null) (NSG) mice in first and secondary recipients. Normal skin melanocytes did not express ErbB4 or EPO-R, but expressed a functional KIT receptor (CD117) as well as NGF-R, ErbB3/Her3, IGF-1-R and CD44. In conclusion, melanoma cells display a unique composition of surface target antigens and cytokine receptors. Malignant transformation of melanomas is accompanied by loss of KIT and acquisition of EPO-R and ErbB4, both of which are co-expressed with NGF-R and PD-1 in distinct subfractions of melanoma cells. However, expression of EPO-R/ErbB4/PD-1 is not indicative of a selective melanoma-initiating potential.


Subject(s)
ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Receptor, Nerve Growth Factor/genetics , Receptors, Erythropoietin/genetics , Skin Neoplasms/genetics , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Line, Tumor , ErbB Receptors/metabolism , Female , Flow Cytometry , Humans , Immunophenotyping , Male , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred NOD , Neoplasm Transplantation , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , Receptor, ErbB-4 , Receptor, Nerve Growth Factor/metabolism , Receptors, Erythropoietin/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
17.
J Immunol ; 191(12): 6147-55, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24227781

ABSTRACT

The growth and differentiation factor activin A is a key regulator of tissue repair, inflammation, fibrosis, and tumorigenesis. However, the cellular targets, which mediate the different activin functions, are still largely unknown. In this study, we show that activin increases the number of mature mast cells in mouse skin in vivo. To determine the relevance of this finding for wound healing and skin carcinogenesis, we mated activin transgenic mice with CreMaster mice, which are characterized by Cre recombinase-mediated mast cell eradication. Using single- and double-mutant mice, we show that loss of mast cells neither affected the stimulatory effect of overexpressed activin on granulation tissue formation and reepithelialization of skin wounds nor its protumorigenic activity in a model of chemically induced skin carcinogenesis. Furthermore, mast cell deficiency did not alter wounding-induced inflammation and new tissue formation or chemically induced angiogenesis and tumorigenesis in mice with normal activin levels. These findings reveal that mast cells are not major targets of activin during wound healing and skin cancer development and also argue against nonredundant functions of mast cells in wound healing and skin carcinogenesis in general.


Subject(s)
Activins/pharmacology , Carcinoma, Squamous Cell/pathology , Mast Cells/physiology , Papilloma/pathology , Skin Neoplasms/pathology , Wound Healing/drug effects , 9,10-Dimethyl-1,2-benzanthracene , Activins/administration & dosage , Activins/deficiency , Animals , Carcinogens , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/chemically induced , Chemotaxis/drug effects , Female , Humans , Injections, Intralesional , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neovascularization, Pathologic/pathology , Neutrophil Infiltration , Papilloma/blood supply , Papilloma/chemically induced , Proto-Oncogene Proteins c-kit/deficiency , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Skin/injuries , Skin/pathology , Skin Neoplasms/blood supply , Skin Neoplasms/chemically induced , Tetradecanoylphorbol Acetate
18.
Blood ; 122(22): 3659-65, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24030387

ABSTRACT

To investigate the role of mast cells in hematopoietic cell transplantation, we assessed graft-versus-host disease (GVHD) in C57BL/6-Kit(W-sh/W-sh) recipients, which virtually lack mast cells, compared with C57BL/6 WT recipients. GVHD was severely exacerbated in C57BL/6-Kit(W-sh/W-sh) mice (median survival time = 13 vs 60 days in wild-type [WT] mice; P < .0001). The increased mortality risk in C57BL/6-Kit(W-sh/W-sh) hosts correlated with increased T-cell numbers in lymph nodes, liver, and gastrointestinal tract sites, as indicated by bioluminescence imaging (P < .001). We did not detect any deficit in the number or function of CD4(+)CD25(+) regulatory T cells (Tregs) in C57BL/6-Kit(W-sh/W-sh) mice. Furthermore, Tregs were equally effective at reducing GVHD in C57BL/6-Kit(W-sh/W-sh) recipients compared with WT recipients containing mast cells. Furthermore, we found that survival of C57BL/6-Kit(W-sh/W-sh) mice during GVHD was significantly improved if the mice were engrafted with bone marrow-derived cultured mast cells from WT C57BL/6 mice but not from interleukin (IL)-10-deficient C57BL/6 mice. These data indicate that the presence of mast cells can significantly reduce GVHD independently of Tregs, by decreasing conventional T-cell proliferation in a mechanism involving IL-10. These experiments support the conclusion that mast cells can mediate a novel immunoregulatory role during hematopoietic cell transplantation.


Subject(s)
Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Mast Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Proliferation , Cell Survival , Female , Graft vs Host Disease/pathology , Hematopoietic Stem Cell Transplantation/adverse effects , Immune Tolerance , Interleukin-10/biosynthesis , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Male , Mast Cells/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/immunology , T-Lymphocytes, Regulatory/classification
19.
J Immunol ; 190(11): 5534-44, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23636054

ABSTRACT

Mast cell-deficient Kit(W-sh) "sash" mice are widely used to investigate mast cell functions. However, mutations of c-Kit also affect additional cells of hematopoietic and nonimmune origin. In this study, we demonstrate that Kit(W-sh) causes aberrant extramedullary myelopoiesis characterized by the expansion of immature lineage-negative cells, common myeloid progenitors, and granulocyte/macrophage progenitors in the spleen. A consistent feature shared by these cell types is the reduced expression of c-Kit. Populations expressing intermediate and high levels of Ly6G, a component of the myeloid differentiation Ag Gr-1, are also highly expanded in the spleen of sash mice. These cells are able to suppress T cell responses in vitro and phenotypically and functionally resemble myeloid-derived suppressor cells (MDSC). MDSC typically accumulate in tumor-bearing hosts and are able to dampen immune responses. Consequently, transfer of MDSC from naive sash mice into line 1 alveolar cell carcinoma tumor-bearing wild-type littermates leads to enhanced tumor progression. However, although it can also be observed in sash mice, accelerated growth of transplanted line 1 alveolar cell carcinoma tumors is a mast cell-independent phenomenon. Thus, the Kit(W-sh) mutation broadly affects key steps in myelopoiesis that may have an impact on mast cell research.


Subject(s)
Mast Cells/immunology , Mutation , Myeloid Cells/immunology , Myelopoiesis/genetics , Myelopoiesis/immunology , Proto-Oncogene Proteins c-kit/genetics , Spleen/cytology , Adoptive Transfer , Animals , Antigens, Ly/metabolism , CD11b Antigen/metabolism , Female , Hematopoiesis, Extramedullary/genetics , Hematopoiesis, Extramedullary/immunology , Immunophenotyping , Mast Cells/metabolism , Mice , Mice, Knockout , Myeloid Cells/metabolism , Neoplasm Transplantation , Neoplasms/immunology , Neoplasms/pathology , Proto-Oncogene Proteins c-kit/deficiency , Spleen/immunology , Spleen/metabolism
20.
Cell Death Dis ; 4: e462, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23328669

ABSTRACT

Various physiologically relevant processes are regulated by the interaction of the receptor tyrosine kinase (c-Kit) and its ligand stem cell factor (SCF), with SCF known to be the most important growth factor for mast cells (MCs). In spite of their traditional role in allergic disorders and innate immunity, MCs have lately emerged as versatile modulators of a variety of physiologic and pathologic processes. Here we show that MCs are critical for pregnancy success. Uterine MCs presented a unique phenotype, accumulated during receptivity and expanded upon pregnancy establishment. Kit(W-sh/W-sh) mice, whose MC deficiency is based on restricted c-Kit gene expression, exhibited severely impaired implantation, which could be completely rescued by systemic or local transfer of wild-type bone marrow-derived MCs. Transferred wild-type MCs favored normal implantation, induced optimal spiral artery remodeling and promoted the expression of MC proteases, transforming growth factor-ß and connective tissue growth factor. MCs contributed to trophoblast survival, placentation and fetal growth through secretion of the glycan-binding protein galectin-1. Our data unveil unrecognized roles for MCs at the fetomaternal interface with critical implications in reproductive medicine.


Subject(s)
Mast Cells/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Animals , Connective Tissue Growth Factor/metabolism , Female , Galectin 1/deficiency , Galectin 1/genetics , Galectin 1/metabolism , Mast Cells/transplantation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , Stem Cell Factor/metabolism , Transforming Growth Factor beta/metabolism , Uterus/anatomy & histology
SELECTION OF CITATIONS
SEARCH DETAIL
...