Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Viruses ; 13(10)2021 09 26.
Article in English | MEDLINE | ID: mdl-34696365

ABSTRACT

Nascent HIV-1 particles incorporate the viral envelope glycoprotein and multiple host transmembrane proteins during assembly at the plasma membrane. At least some of these host transmembrane proteins on the surface of virions are reported as pro-viral factors that enhance virus attachment to target cells or facilitate trans-infection of CD4+ T cells via interactions with non-T cells. In addition to the pro-viral factors, anti-viral transmembrane proteins are incorporated into progeny virions. These virion-incorporated transmembrane proteins inhibit HIV-1 entry at the point of attachment and fusion. In infected polarized CD4+ T cells, HIV-1 Gag localizes to a rear-end protrusion known as the uropod. Regardless of cell polarization, Gag colocalizes with and promotes the virion incorporation of a subset of uropod-directed host transmembrane proteins, including CD162, CD43, and CD44. Until recently, the functions of these virion-incorporated proteins had not been clear. Here, we review the recent findings about the roles played by virion-incorporated CD162, CD43, and CD44 in HIV-1 spread to CD4+ T cells.


Subject(s)
HIV Infections/metabolism , Hyaluronan Receptors/metabolism , Leukosialin/metabolism , Membrane Glycoproteins/metabolism , Cell Membrane/metabolism , HIV Infections/genetics , HIV-1/genetics , HIV-1/metabolism , HIV-1/pathogenicity , Host-Pathogen Interactions , Humans , Hyaluronan Receptors/genetics , Leukosialin/genetics , Membrane Glycoproteins/genetics , Membrane Proteins/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Virion/metabolism , Virus Assembly , Virus Attachment , gag Gene Products, Human Immunodeficiency Virus/metabolism
2.
Proc Natl Acad Sci U S A ; 117(14): 8055-8063, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32193343

ABSTRACT

HIV-1 particles incorporate various host transmembrane proteins in addition to viral Env glycoprotein during assembly at the plasma membrane. In polarized T cells, HIV-1 structural protein Gag localizes to the plasma membrane of uropod, a rear-end protrusion. Notably, uropod transmembrane proteins PSGL-1 and CD43 cocluster specifically with Gag assembling at the plasma membrane even in cells that do not form uropods. Recent reports have shown that expression of either PSGL-1 or CD43 in virus-producing cells reduces the infectivity of progeny virions and that HIV-1 infection reduces the cell surface expression of these proteins. However, the mechanisms for both processes remain to be determined. In this study, we found that virion incorporation of PSGL-1 and CD43 closely correlates with diminished virion infectivity. PSGL-1 and CD43 inhibited virus attachment to CD4+ cells irrespective of the presence of Env. These proteins also inhibited virion attachment to CD4- lymphoid organ fibroblastic reticular cells that mediate transinfection of CD4+ T cells. Consistent with the possibility that highly extended extracellular domains of these proteins physically block virus-cell attachment, the inhibitory effect of PSGL-1 required its full-length ectodomain. HIV-1 encoding Gag mutants that are defective in either coclustering with these host proteins or ESCRT-dependent particle release failed to reduce PSGL-1 on surface of infected cells. This study reveals an anti-HIV-1 mechanism that suppresses virus-cell attachment and a previously unappreciated process of HIV-1-mediated down-regulation of host antiviral proteins, both of which likely require virion incorporation of these proteins.


Subject(s)
HIV Infections/immunology , HIV-1/immunology , Host-Pathogen Interactions/genetics , Leukosialin/genetics , Membrane Glycoproteins/genetics , Virion/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism , Blood Buffy Coat/cytology , Down-Regulation , Gene Knockout Techniques , HEK293 Cells , HIV Infections/virology , HIV-1/genetics , HIV-1/metabolism , HeLa Cells , Healthy Volunteers , Host-Pathogen Interactions/immunology , Humans , Mutation , Protein Domains/genetics , T-Lymphocytes/immunology , Virus Assembly/genetics , Virus Assembly/immunology , Virus Attachment , Virus Replication/genetics , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology
3.
Stem Cells Dev ; 28(23): 1540-1551, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31595840

ABSTRACT

Red blood cell (RBC) differentiation from human induced pluripotent stem cells (hiPSCs) offers great potential for developmental studies and innovative therapies. However, ex vivo erythropoiesis from hiPSCs is currently limited by low efficiency and unphysiological conditions of common culture systems. Especially, the absence of a physiological niche may impair cell growth and lineage-specific differentiation. We here describe a simplified, xeno- and feeder-free culture system for prolonged RBC generation that uses low numbers of supporting cytokines [stem cell factor (SCF), erythropoietin (EPO), and interleukin 3 (IL-3)] and is based on the intermediate development of a "hematopoietic cell forming complex (HCFC)." From this HCFC, CD43+ hematopoietic cells (purity >95%) were continuously released into the supernatant and could be collected repeatedly over a period of 6 weeks for further erythroid differentiation. The released cells were mainly CD34+/CD45+ progenitors with high erythroid colony-forming potential and CD36+ erythroid precursors. A total of 1.5 × 107 cells could be harvested from the supernatant of one six-well plate, showing 100- to 1000-fold amplification during subsequent homogeneous differentiation into GPA+ erythroid cells. Mean enucleation rates near 40% (up to 60%) further confirmed the potency of the system. These benefits may be explained by the generation of a niche within the HCFC that mimics the spatiotemporal signaling of the physiological microenvironment in which erythropoiesis occurs. Compared to other protocols, this method provides lower complexity, less cytokine and medium consumption, higher cellular output, and better enucleation. In addition, slight modifications in cytokine addition shift the system toward continuous generation of granulocytes and macrophages.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/genetics , Erythroid Cells/cytology , Induced Pluripotent Stem Cells/cytology , CD36 Antigens/genetics , Cell Lineage/genetics , Cellular Microenvironment/genetics , Cytokines/genetics , Erythrocytes/cytology , Erythropoiesis/genetics , Hematopoietic Stem Cells/cytology , Humans , Leukosialin/genetics
4.
Cytometry B Clin Cytom ; 96(5): 417-425, 2019 09.
Article in English | MEDLINE | ID: mdl-31157955

ABSTRACT

BACKGROUND: Despite bone marrow (BM) immunophenotyping by flow cytometry has progressively been recognized as an important tool for the diagnosis of myelodysplastic syndromes (MDS), the sparse knowledge about normal erythroid maturation and the lack of markers for erythroid characterization is a major shortcoming. METHODS: Here, we analyzed the expression of CD43 and CD49d, two markers included in the diagnostic panel for B-cell chronic lymphoproliferative disorders (B-CLPD), in the CD34+ compartment of normal BM and along the normal and dysplastic erythroid maturation. For this, 13 normal BM aspirates and 18 BM aspirates from MDS patients were studied by flow cytometry. RESULTS: Normal BM presented a higher expression of CD43 and CD49d among CD34+ erythroid precursors, compared to CD34+ cells committed to the remaining hematopoietic cell lineages. CD43 expression progressively decreased along the normal erythroid maturation, whereas CD49d levels increased from Stage I to Stage II, were maintained in Stages II and III, and then decreased until the last stage of maturation. In MDS, the expression of CD43 and CD49d followed a similar pattern, but with decreased expression levels for both markers, observed in all erythroid maturation stages (P < 0.05). CONCLUSIONS: Our results point to the usefulness of CD43 and CD49d, two markers commonly present in B-CLPD diagnosis panels, in the identification of dysplastic phenotypic features in the erythroid lineage. This allows a feasible and inexpensive way to identify patients who would benefit from a more extensive study to evaluate the presence of MDS, during the processing of suspected B-CLPD samples. © 2019 International Clinical Cytometry Society.


Subject(s)
Integrin alpha4/genetics , Leukosialin/genetics , Lymphoproliferative Disorders/diagnosis , Myelodysplastic Syndromes/diagnosis , Aged , Aged, 80 and over , Antigens, CD34/immunology , B-Lymphocytes/pathology , Biomarkers, Tumor/immunology , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Lineage/immunology , Female , Flow Cytometry/methods , Humans , Immunophenotyping/methods , Integrin alpha4/immunology , Leukosialin/immunology , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/pathology , Male , Middle Aged , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/pathology
5.
Cytometry B Clin Cytom ; 96(4): 275-282, 2019 07.
Article in English | MEDLINE | ID: mdl-31077558

ABSTRACT

BACKGROUND: Hairy cell leukemia (HCL) and hairy cell leukemia variant (HCLv) are rare diseases with overlapping clinicopathological features. Features distinguishing HCL from HCLv include expression of CD25, CD123, CD200, annexin-A1, and the presence of BRAF V600E mutation. HCLv typically lacks these markers, but they may occur in a subgroup of HCL patients with an aggressive clinical course. We examined CD43, CD81, CD79b, and CD200 expression in HCL and HCLv. METHODS: Multiparametric flow cytometry (FCM) was performed on blood from 59 HCL and 15 HCLv patients for protocol entry. Mean fluorescent intensity (MFI) of CD43, CD79b, CD81, and CD200 was determined (for CD200, n = 17 and 7, respectively). RESULTS: Median MFI of HCL vs HCLv was 545 vs 272 for CD43, 602 vs 2,450 for CD81, 4,962 vs 1,969 for CD79b, and 11,652 vs 1,405 for CD200, respectively. Analysis of the median differences, HCL minus HCLv (and their 95% confidence intervals and P-values) indicated that CD43 MFI (estimated median difference (95% CI): 212 [72-413; P = 0.0027) and CD200 MFI (9,883 [3,514-13,434]; P < 0.0001) were higher in HCL than in HCLv, while CD81 MFI (-1,858 [-2,604 to -1,365]; P < 0.0001) was lower in HCL than in HCLv. CD79b MFI HCL median was more than double that of HCLv, but the observed difference (1,571 [-739 to 4,417]) was consistent with the null hypothesis of no difference (P = 0.13). CONCLUSIONS: CD200, CD43, and CD81 are likely differentially expressed between HCL and HCLv, reflecting their differing disease biology. Inclusion of these markers in FCM is potentially informative. © 2019 International Clinical Cytometry Society.


Subject(s)
Antigens, CD/genetics , Leukemia, Hairy Cell/genetics , Leukosialin/genetics , Tetraspanin 28/genetics , Adult , Aged , Aged, 80 and over , Female , Flow Cytometry , Genetic Variation/genetics , Humans , Immunophenotyping , Leukemia, Hairy Cell/diagnosis , Male , Middle Aged
6.
Immunology ; 157(1): 52-69, 2019 05.
Article in English | MEDLINE | ID: mdl-30690734

ABSTRACT

T helper type 17 lymphocytes (Th17 cells) infiltrate the central nervous system (CNS), induce inflammation and demyelination and play a pivotal role in the pathogenesis of multiple sclerosis. Sialomucin CD43 is highly expressed in Th17 cells and mediates adhesion to endothelial selectin (E-selectin), an initiating step in Th17 cell recruitment to sites of inflammation. CD43-/- mice have impaired Th17 cell recruitment to the CNS and are protected from experimental autoimmune encephalomyelitis (EAE), the mouse model of multiple sclerosis. However, E-selectin is dispensable for the development of EAE, in contrast to intercellular and vascular cell adhesion molecules (ICAM-1 and VCAM-1). We report that CD43-/- mice have decreased demyelination and T-cell infiltration, but similar up-regulation of ICAM-1 and VCAM-1 in the spinal cord, compared with wild-type (WT) mice, at the initiation of EAE. CD43-/- Th17 cells have impaired adhesion to ICAM-1 under flow conditions in vitro, despite having similar expression of LFA-1, the main T-cell ligand for ICAM-1, as WT Th17 cells. Regardless of the route of integrin activation, CD43-/- Th17 cell firm arrest on ICAM-1 was comparable to that of WT Th17 cells, but CD43-/- Th17 cells failed to optimally apically migrate on immobilized ICAM-1-coated coverslips and endothelial cells, and to transmigrate under shear flow conditions in an ICAM-1-dependent manner. Collectively, these findings unveil novel roles for CD43, facilitating adhesion of Th17 cells to ICAM-1 and modulating apical and transendothelial migration, as mechanisms potentially responsible for Th17 cell recruitment to sites of inflammation such as the CNS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/immunology , Intercellular Adhesion Molecule-1/metabolism , Leukosialin/metabolism , Multiple Sclerosis/immunology , Th17 Cells/immunology , Animals , Cell Adhesion , Cell Movement , Disease Models, Animal , Humans , Intercellular Adhesion Molecule-1/genetics , Leukosialin/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Transendothelial and Transepithelial Migration , Vascular Cell Adhesion Molecule-1/metabolism
7.
J Proteome Res ; 18(3): 1125-1132, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30582698

ABSTRACT

C-type lectins are a diverse group of proteins involved in many human physiological and pathological processes. Most C-type lectins are glycan-binding proteins, some of which are pivotal for innate immune responses against pathogens. Other C-type lectins, such as the macrophage galactose-type lectin (MGL), have been shown to induce immunosuppressive responses upon the recognition of aberrant glycosylation on cancer cells. MGL is known to recognize terminal N-acetylgalactosamine (GalNAc), such as the Tn antigen, which is commonly found on malignant cells. Even though this glycan specificity of MGL is well described, there is a lack of understanding of the actual glycoproteins that bind MGL. We present a glycoproteomic workflow for the identification of MGL-binding proteins, which we applied to study MGL ligands on the human Jurkat leukemia cell line. In addition to the known MGL ligands and Tn antigen-carrying proteins CD43 and CD45 on these cells, we have identified a set of novel cell-surface ligands for MGL. Importantly, for several of these, O-glycosylation has hitherto not been described. Altogether, our data provide new insight into the identification and structure of novel MGL ligands that presumably act as modulatory molecules in cancer immune responses.


Subject(s)
Glycoproteins/genetics , Lectins, C-Type/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Acetylgalactosamine/genetics , Acetylgalactosamine/metabolism , Antigens, Tumor-Associated, Carbohydrate/genetics , Antigens, Tumor-Associated, Carbohydrate/metabolism , Carrier Proteins/genetics , Carrier Proteins/immunology , Glycoproteins/immunology , Glycosylation , Humans , Immunity, Innate/genetics , Jurkat Cells , Lectins, C-Type/immunology , Leukocyte Common Antigens/genetics , Leukosialin/genetics , Ligands , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology
8.
Biochem Biophys Res Commun ; 506(1): 60-65, 2018 11 17.
Article in English | MEDLINE | ID: mdl-30336974

ABSTRACT

Investigations on the structure and functional roles of glycosylation - an intricate, complex, and dynamic post translational modification on proteins - in biological processes has been a challenging task. Glycan modifications vary depending on the specific cell type, its developmental stage, and resting or activated state. In the present study, we aim to understand the differences between the mucin-type O-glycosylation (MTOG) of two functionally divergent human cell lines, K562 (chronic myeloid leukemia) and U937 (histiocytic lymphoma), having myeloid origins. MTOG is initiated by the addition of N-acetyl-α-d-galactosamine (GalNAc) to Ser/Thr of glycoproteins. We exploited the metabolic glycan engineering (MGE) strategy using the peracetyl N-thioglycolyl-d-galactosamine (Ac5GalNTGc), a synthetic GalNAc analogue, to engineer the glycoconjugates. Ac5GalNTGc was metabolized and incorporated as N-thioglycolyl-d-galactosamine (GalNTGc) in cell surface glycoproteins in both the cell lines with varying degrees of efficiency. Notably, metabolic incorporation of GalNTGc resulted in differential inhibition of MTOG. It was observed that endogenous glycosylation machinery of K562 is relatively more stringent for selecting GalNTGc whereas U937 is flexible towards this selection. Additionally, we studied how the glycan modifications vary on a given CD antigen in these cell lines. Particularly, MTOG on CD43 was differentially inhibited in K562 and U937 as revealed by glycan-dependent and glycan-independent antibodies. It was observed that the effect of MGE on CD43 was similar to global effects on both cell lines. Consequences of MGE using GalNAc analogues depend on the expression and activity of various glycosyl transferases which determine global glycosylation on cell surface as well as on specific glycoproteins.


Subject(s)
Acetylgalactosamine/metabolism , Glycoconjugates/metabolism , Glycoproteins/metabolism , Leukosialin/metabolism , Mucins/metabolism , Protein Processing, Post-Translational , Acetylgalactosamine/chemistry , Cell Line, Tumor , Gene Expression , Glycoconjugates/chemistry , Glycoproteins/chemistry , Glycoproteins/genetics , Glycosylation , Humans , Jurkat Cells , K562 Cells , Leukosialin/chemistry , Leukosialin/genetics , Metabolic Engineering , Monocytes/cytology , Monocytes/metabolism , Mucins/chemistry , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Organ Specificity
9.
PLoS One ; 13(9): e0202656, 2018.
Article in English | MEDLINE | ID: mdl-30226896

ABSTRACT

CD43 is a large transmembrane protein involved in T cell activation. Previous studies of CD43-/- mice in viral models have demonstrated a role for CD43 in Th1/Th2 skewing, activation of Foxp3+ Treg, and T cell apoptosis. However, the role of CD43 during sepsis has never been tested. Thus, we interrogated the role of CD43 during sepsis using a murine cecal ligation and puncture (CLP) model, and found that CD43-/- mice demonstrated significantly worsened mortality compared to B6 mice following CLP. Phenotypic analysis of splenocytes isolated 24 h after septic insult revealed significantly increased apoptosis of central memory cells in both CD4+ and CD8+ T cell compartments in CD43-/- septic mice compared to WT septic mice. Furthermore, CD43-/-septic mice exhibited a prominent Th2 skewing following sepsis relative to WT septic mice, as evidenced by a significant decrease in the frequency of IL-2+ CXCR3+ TH1 cells as a significant increase in the frequency of IL-4+ CCR4+ TH2 cells. Finally, septic CD43-/- animals contained significantly fewer CD25+ Foxp3+ TReg cells as compared to WT septic animals. Importantly, depleting CD25+ Treg eliminated the increased mortality observed in CD43-/- mice. Taken together, these data demonstrate an important role of CD43 in modulating immune dysregulation and mortality following sepsis.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Leukosialin/genetics , Sepsis/mortality , Spleen/immunology , Animals , Apoptosis , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Gene Knockout Techniques , Interleukins/metabolism , Male , Mice , Receptors, CXCR/metabolism , Sepsis/genetics , Sepsis/immunology , Spleen/cytology , Th1 Cells/cytology , Th1 Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology
10.
Cancer Med ; 7(9): 4284-4295, 2018 09.
Article in English | MEDLINE | ID: mdl-30019388

ABSTRACT

Both CD5 and CD43 are expressed on the surface of B lymphocytes of definite phase and associated with the adverse outcome in diffuse large B-cell lymphoma (DLBCL). However, the relationship between CD5 and CD43 expression and the prognostic value of CD5/CD43 coexpression in DLBCL are unknown. We herein determined the correlation between CD5 and CD43 expression, as separate factors or in combination, with the clinicopathological features and survival of 200 patients with DLBCL receiving standard chemotherapy with or without rituximab. Among these DLBCL patients, CD5 expression, CD43 expression, and CD5/CD43 coexpression were detected in 18 (9%), 57 (27%), and 10 (5%) patients, respectively, and all were positively correlated with advanced age and nongerminal cell type. CD5-positive and CD43-positive DLBCL patients had poorer event-free survival (EFS, P < 0.001) and overall survival (OS, P < 0.001) than CD5-negative and CD43-negative patients, respectively. CD5/CD43 coexpression was correlated with a significantly worse prognosis than CD5 or CD43 expression alone. Univariate analysis showed that CD5 expression, CD43 expression, and CD5/CD43 coexpression were all adverse prognostic factors for DLBCL patient survival, and CD5/CD43 coexpression was associated with a greater relative risk for recurrence and death than either CD5 or CD43 expression alone. Multivariate analysis demonstrated that CD5/CD43 coexpression was an independent prognostic factor for EFS (P < 0.001) and OS (P < 0.001) in DLBCL. In conclusion, our data indicate that DLBCL patients with CD5/CD43 coexpression represent a specific subgroup with a significantly worse prognosis than those expressing either marker alone.


Subject(s)
Biomarkers, Tumor , CD5 Antigens/metabolism , Leukosialin/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/mortality , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CD5 Antigens/genetics , Child , Female , Gene Expression , Gene Expression Profiling , Humans , Immunohistochemistry , Immunophenotyping , Leukosialin/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/therapy , Male , Middle Aged , Prognosis , Treatment Outcome , Young Adult
11.
Stem Cell Rev Rep ; 14(4): 525-534, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29520567

ABSTRACT

Identification of transcription factors that directly convert pluripotent stem cells (PSCs) into endothelial and blood cells and advances in the chemical modifications of messenger RNA (mRNA) offer alternative nucleic acid-based transgene-free approach for scalable production of these cells for drug screening and therapeutic purposes. Here we evaluated the effect of 5' and 3' RNA untranslated regions (UTRs) on translational efficiency of chemically-modified synthetic mRNA (modRNA) in human PSCs and showed that an addition of 5'UTR indeed enhanced protein expression. With the optimized modRNAs expressing ETV2 or ETV2 and GATA2, we are able to produce VE-cadherin+ endothelial cells and CD34+CD43+ hematopoietic progenitors, respectively, from human PSCs as well as non-human primate (NHP) PSCs. Overall, our findings provide valuable information on the design of in vitro transcription templates being used in PSCs and its broad applicability for basic research, disease modeling, and regenerative medicine.


Subject(s)
Endothelial Cells/metabolism , Hematopoietic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , 5' Untranslated Regions/genetics , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, CD34/genetics , Antigens, CD34/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Differentiation/genetics , Cell Line , Cells, Cultured , Endothelial Cells/cytology , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Hematopoietic Stem Cells/cytology , Humans , Leukosialin/genetics , Leukosialin/metabolism , Pluripotent Stem Cells/cytology , Primates , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Stem Cells Dev ; 27(8): 524-533, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29583085

ABSTRACT

Specification of endothelial cells (ECs) into arterial, venous, and lymphatic cells is a crucial process of vascular development, and expanding our knowledge about EC specification from human pluripotent stem cells (hPSCs) will aid the design of optimal strategies for producing desired types of ECs for therapies. In our prior studies, we revealed that hPSC-derived VE-cadherin(V)+CD31+CD34+ ECs are heterogeneous and include at least three major subsets with distinct hemogenic properties: V+CD43/235a-CD73- hemogenic endothelial progenitors (HEPs), V+CD43loCD235a+73- angiogenic hematopoietic progenitors (AHPs), and V+CD43/235a-73+ non-HEPs. In this study, using angiogenesis assays, we demonstrated that ECs within these subsets have distinct endothelial colony- and tube-forming properties, proliferative and migratory properties, and endothelial nitric oxide synthase and inflammatory cytokine production potentials. Culture of isolated subsets in arterial, venous, and lymphatic conditions revealed that AHPs are skewed toward lymphatic, HEPs toward arterial, and non-HEPs toward venous differentiation in vitro. These findings suggest that selection and enhancement of production of a particular EC subset may aid in generating desirable EC populations with arterial, venous, or lymphatic properties from hPSCs.


Subject(s)
Cell Lineage/physiology , Hemangioblasts/cytology , Neovascularization, Physiologic , Pluripotent Stem Cells/cytology , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, CD34/genetics , Antigens, CD34/metabolism , Biomarkers/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Differentiation , Cell Line , Cell Movement , Cell Proliferation , Colony-Forming Units Assay , Cytokines/genetics , Cytokines/metabolism , Gene Expression , Hemangioblasts/physiology , Humans , Leukosialin/genetics , Leukosialin/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pluripotent Stem Cells/physiology
13.
Cell Death Dis ; 9(3): 293, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29463785

ABSTRACT

Regulatory T cells in rejected allograft patients display an inability to control responder T cells. Galectin-1 (Gal1) inhibits responder T cells through binding CD7. We investigated whether the dysfunctional immunoregulation in liver allograft rejection patients results from reduced regulatory T-cell Gal1 expression and/or responder T-cell CD7 expression. Circulating regulatory T cells and responder T cells were profiled from 31 acute rejection transplant patients, 85 transplant patients in remission, and 40 healthy controls. CD7+ and CD7- responder T cells were co-cultured with regulatory T cells to assess regulatory T-cell suppressor function. Gal1-small interfering RNA was used to silence regulatory T-cell Gal1. The CD7+ cell percentage was inversely correlated with AST, ALT, and GGT levels. The proportions of CD7+ responder T cells and Gal1+ regulatory T cells were higher in healthy controls than in transplant patients in remission and lowest in acute rejection transplant patients. Notably, CD7+ responder T-cell susceptibility to Gal1+ regulatory T-cell control was ranked in the same manner. Silencing Gal1 expression in regulatory T cells reduced their ability to suppress CD7+ (but not CD7-) responder T cells. Additionally, the proportions of CD43+ and CD45+ responder T cells were higher in healthy controls than in acute rejection transplant patients. CD43 co-expression (but not CD45 co-expression) on CD7+ responder T cells promoted their apoptosis in a Gal1-dependent manner. In sum, dysfunctional immunoregulation in liver allograft rejection patients can be partly attributed to reduced regulatory T-cell Gal1 expression and reduced responder T-cell CD7 expression. Responder T-cell CD43 downregulation in acute rejection patients may further contribute to reduced responder T-cell responsiveness to regulatory T-cell control.


Subject(s)
Allografts/immunology , Antigens, CD7/immunology , Galectin 1/immunology , Graft Rejection/immunology , Liver/surgery , Adult , Aged , Antigens, CD7/genetics , Apoptosis , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/physiopathology , Carcinoma, Hepatocellular/surgery , Female , Galectin 1/genetics , Graft Rejection/genetics , Humans , Leukosialin/genetics , Leukosialin/immunology , Liver/immunology , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/physiopathology , Liver Neoplasms/surgery , Liver Transplantation , Male , Middle Aged , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Young Adult
14.
Stem Cell Res ; 17(2): 401-405, 2016 09.
Article in English | MEDLINE | ID: mdl-27879215

ABSTRACT

Human embryonic stem cell line WA01 was genetically modified using zinc-finger nucleases and the PiggyBac/transponson system to introduce a fluorescence reporter for VE-cadherin (VEC; tdTomato) and CD43 (eGFP). Phenotypic and functional assays for pluripotency revealed the modified hES cell reporter lines remained normal. When the cells were differentiated into hematoendothelial lineages, either by directed differentiation or direct reprogramming, flow cytometric and fluorescence microscopy showed that VEC+ endothelial cells express tdTomato and CD43+ hematopoietic progenitors express eGFP.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/cytology , Leukosialin/metabolism , Antigens, CD/genetics , Cadherins/genetics , Cell Differentiation , Cells, Cultured , Embryoid Bodies/metabolism , Embryoid Bodies/pathology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Genes, Reporter , Genetic Vectors/genetics , Genetic Vectors/metabolism , Green Fluorescent Proteins/genetics , Hematopoietic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Karyotype , Leukosialin/genetics , Male , Microscopy, Fluorescence , Time-Lapse Imaging , Transcription Factors/genetics , Transcription Factors/metabolism
15.
BMC Med Genet ; 17(Suppl 1): 68, 2016 Oct 10.
Article in English | MEDLINE | ID: mdl-27766954

ABSTRACT

BACKGROUND: Osteoarthritis (OA) is a progressive joint disease characterized by gradual degradation of extracellular matrix (ECM) components in the cartilage and bone. The ECM of cartilage is a highly specified structure that is mainly composed of type II collagen and provides tensile strength to the tissue via aggrecan and proteoglycans. However, changes in the ECM composition and structure can lead to loss of collagen type II and network integrity. Several risk factors have been correlated with OA including age, genetic predisposition, hereditary factors, obesity, mechanical injuries, and joint trauma. Certain genetic association studies have identified several genes associated with OA using genome-wide association studies (GWASs). RESULTS: We identified several novel genetic variants affecting genes that function in several candidate causative pathways including immune responses, inflammatory and cartilage degradation such as SELP, SPN, and COL6A6. CONCLUSIONS: The approach of whole-exome sequencing can be a promising method to identify genetic mutations that can influence the OA disease.


Subject(s)
Exome/genetics , Genetic Variation , Osteoarthritis/genetics , Aged , Cartilage/metabolism , Collagen Type II/genetics , Collagen Type VI/genetics , Genome-Wide Association Study , Humans , Leukosialin/genetics , Middle Aged , Osteoarthritis/pathology , P-Selectin/genetics , Polymorphism, Single Nucleotide , Risk Factors
16.
J Biol Chem ; 290(37): 22662-77, 2015 Sep 11.
Article in English | MEDLINE | ID: mdl-26216879

ABSTRACT

During inflammation, dendritic cells emigrate from inflamed tissue across the lymphatic endothelium into the lymphatic vasculature and travel to regional lymph nodes to initiate immune responses. However, the processes that regulate dendritic cell tissue egress and migration across the lymphatic endothelium are not well defined. The mammalian lectin galectin-1 is highly expressed by vascular endothelial cells in inflamed tissue and has been shown to regulate immune cell tissue entry into inflamed tissue. Here, we show that galectin-1 is also highly expressed by human lymphatic endothelial cells, and deposition of galectin-1 in extracellular matrix selectively regulates migration of specific human dendritic cell subsets. The presence of galectin-1 inhibits migration of immunogenic dendritic cells through the extracellular matrix and across lymphatic endothelial cells, but it has no effect on migration of tolerogenic dendritic cells. The major galectin-1 counter-receptor on both dendritic cell populations is the cell surface mucin CD43; differential core 2 O-glycosylation of CD43 between immunogenic dendritic cells and tolerogenic dendritic cells appears to contribute to the differential effect of galectin-1 on migration. Binding of galectin-1 to immunogenic dendritic cells reduces phosphorylation and activity of the protein-tyrosine kinase Pyk2, an effect that may also contribute to reduced migration of this subset. In a murine lymphedema model, galectin-1(-/-) animals had increased numbers of migratory dendritic cells in draining lymph nodes, specifically dendritic cells with an immunogenic phenotype. These findings define a novel role for galectin-1 in inhibiting tissue emigration of immunogenic, but not tolerogenic, dendritic cells, providing an additional mechanism by which galectin-1 can dampen immune responses.


Subject(s)
Cell Movement/immunology , Dendritic Cells/immunology , Endothelial Cells/immunology , Galectin 1/immunology , Animals , Cell Line , Cell Movement/genetics , Dendritic Cells/pathology , Disease Models, Animal , Endothelial Cells/pathology , Extracellular Matrix/genetics , Extracellular Matrix/immunology , Focal Adhesion Kinase 2/genetics , Focal Adhesion Kinase 2/immunology , Galectin 1/genetics , Glycosylation , Humans , Leukosialin/genetics , Leukosialin/immunology , Lymphedema/genetics , Lymphedema/immunology , Lymphedema/pathology , Mice , Mice, Knockout
17.
Arterioscler Thromb Vasc Biol ; 35(2): 309-11, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25524774

ABSTRACT

OBJECTIVE: The aim of this study was to investigate the role of cluster of differentiation 43 (CD43), an integral membrane glycoprotein with both proadhesive and antiadhesive activities, in atherosclerosis. APPROACH AND RESULTS: Low-density lipoprotein receptor-deficient mice were lethally irradiated and reconstituted with either bone marrow from CD43(-/-) mice or from wild-type controls. We found that mice lacking the CD43 on their leukocytes had significantly less severe atherosclerosis and that, contrary to our expectation, macrophage infiltration into the vessel wall was not affected by the lack of CD43 in the leukocytes. However, we found that CD43 mediates cholesterol homeostasis in macrophages by facilitating cholesterol efflux. This resulted in a significant reduction in storage of cholesterol in the aorta of mice lacking CD43 in the leukocytes. CONCLUSIONS: CD43 may be an important mediator of macrophage lipid homeostasis, thereby affecting macrophage foam cell formation and ultimately atherosclerotic plaque development.


Subject(s)
Aorta/metabolism , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Leukocytes/metabolism , Leukosialin/metabolism , Animals , Aorta/immunology , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Cholesterol/metabolism , Disease Models, Animal , Down-Regulation , Leukocytes/immunology , Leukosialin/genetics , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic , Receptors, LDL/deficiency , Receptors, LDL/genetics , Time Factors
18.
Am J Respir Cell Mol Biol ; 52(3): 342-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25078322

ABSTRACT

CD43, a surface glycoprotein, regulates Mycobacterium tuberculosis macrophage binding, replication, and proinflammatory cytokine induction in a murine model. We hypothesized that single-nucleotide polymorphisms (SNPs) in the CD43 gene region are associated with human tuberculosis (TB) susceptibility. We performed a case-population study in discovery (352 TB cases and 382 control subjects) and validation cohorts (339 TB cases and 376 control subjects). We examined whether 11 haplotype-tagging SNPs in the CD43 gene region were associated with tuberculous meningitis (TBM) and pulmonary TB (PTB) in Vietnam. Three SNPs from the CD43 gene region were associated with TB susceptibility with a genotypic model. The association fit a recessive genetic model and was greater for TBM than for PTB (for TBM: rs4788172, odds ratio [OR], 1.64; 95% confidence interval [CI], 1.04-2.59, rs17842268 [OR, 2.20; 95% CI, 1.29-3.76, and rs12596308 [OR, 2.38; 95% CI, 1.47-3.89]). Among TBM cases, rs17842268 was associated with decreased survival (hazard ratio, 2.7; 95% CI, 1.1-6.5; P = 0.011). In addition, rs12596308 and rs17842268 were associated with focal neurologic deficit at TBM presentation. Our data suggest that CD43 polymorphisms are associated with TB susceptibility, disease manifestations, and worse outcomes. To our knowledge, this is the first report that links CD43 genetic variants with susceptibility and outcome from a disease.


Subject(s)
Genetic Predisposition to Disease/genetics , Leukosialin/genetics , Polymorphism, Single Nucleotide/genetics , Tuberculosis, Meningeal/genetics , Tuberculosis, Pulmonary/genetics , Case-Control Studies , Haplotypes/genetics , Humans , Mycobacterium tuberculosis
19.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 31(5): 561-4, 2014 Oct.
Article in Chinese | MEDLINE | ID: mdl-25297581

ABSTRACT

OBJECTIVE: To investigate the spectrum of ß -thalassemia mutations in Guizhou Province. METHODS: For 542 individuals suspected to have ß -thalassemia by decreased mean corpuscular volume (MCV) and corpuscle hemoglobin (MCH) by routine blood test and hemoglobin electrophoresis, reverse dot blot hybridization (RDB) was performed to detect 17 known ß -thalassemia mutations, including 8 common and 9 rare mutations. For cases where no mutation was identified, the entire human ß -globin gene was screened to find other rare mutations. The distribution and frequencies of detected ß -thalassemia mutations were then analyzed. RESULTS: A total of 460 individuals were diagnosed as ß -thalassemia by DNA analysis, which included 352 heterozygotes, 67 compound heterozygotes and 41 mutant homozygotes. A total of 12 ß -thalassemia mutations were detected in these individuals. The mutations have ranked from high to low frequency as: CD17 (40.74%), CD41-42 (33.69%), IVS-II-654 (13.76%), -28 (3.70%), ß E (3.35%), CD71-72(1.94%), CD43 (1.06%), IVS-I-1 (0.71%), CD27-28 (0.35%), -29(0.35%), CAP (0.18%), and CD121 (0.18%). The former six mutations have accounted for 97.18% of all. CD121 (GAA> TAA) detected from a heterozygote, as a dominant mutation, has been firstly found in the Chinese population. CONCLUSION: The spectrum of ß -thalassemia in Guizhou Province showed certain distinct characteristics, with CD17 being the most common mutation. The newly discovered mutation of CD121 has expanded the spectrum of ß -thalassemia in Chinese population. Our result may provide valuable information for the prevention and control of ß -thalassemia in Guizhou.


Subject(s)
Mutation , beta-Thalassemia/genetics , Adolescent , Adult , Asian People/genetics , Child , Child, Preschool , China , DNA Mutational Analysis , Female , Humans , Infant , Leukosialin/genetics , Male , Middle Aged , Platelet Membrane Glycoprotein IIb/genetics , Receptors, Interleukin-1 Type I/genetics , Young Adult , beta-Globins/genetics , beta-Thalassemia/diagnosis , beta-Thalassemia/ethnology
20.
Stem Cell Reports ; 3(3): 489-501, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25241746

ABSTRACT

Definitive hematopoietic stem cells (HSCs) develop in the aorta gonad mesonephros (AGM) region in a stepwise manner. Type I pre-HSCs express CD41 but lack CD45 expression, which is subsequently upregulated in type II pre-HSCs prior to their maturation into definitive HSCs. Here, using ex vivo modeling of HSC development, we identify precursors of definitive HSCs in the trunk of the embryonic day 9.5 (E9.5) mouse embryo. These precursors, termed here pro-HSCs, are less mature than type I and II pre-HSCs. Although pro-HSCs are CD41(+), they lack the CD43 marker, which is gradually upregulated in the developing HSC lineage. We show that stem cell factor (SCF), but not interleukin-3 (IL-3), is a major effector of HSC maturation during E9-E10. This study extends further the previously established hierarchical organization of the developing HSC lineage and presents it as a differentially regulated four-step process and identifies additional targets that could facilitate the generation of transplantable HSCs from pluripotent cells for clinical needs.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells/cytology , Interleukin-3/metabolism , Leukosialin/genetics , Stem Cell Factor/metabolism , Animals , Aorta/embryology , Cell Lineage , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Gene Deletion , Hematopoietic Stem Cells/metabolism , Mesonephros/embryology , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...