Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Aging (Albany NY) ; 12(20): 19867-19879, 2020 10 16.
Article in English | MEDLINE | ID: mdl-33065551

ABSTRACT

The ongoing outbreak of COVID-19 has been announced by the World Health Organization as a worldwide public health emergency. The aim of this study was to distinguish between severe and non-severe patients in early diagnosis. The results showed that the mortality of COVID-19 patients increased accompanied by age. Host factors CRP, IL-1ß, hs-CRP, IL-8, and IL-6 levels in severe pneumonia patients were higher than in non-severe patients. CD3, CD8, and CD45 counts were decreased in COVID-19 patients. The results of this study suggest that the K-values of CD45 might be useful in distinguishing between severe and non-severe cases. The cut-off value for CD45 was -94.33. The K-values for CD45 in non-severe case were above the cut-off values, indicating a 100% prediction success rate for severe and non-severe cases following SARS-CoV-2 infection. The results confirmed that immune system dysfunction is a potential cause of mortality following COVID-19 infection, particularly for the elderly. CD45 deficiency dysfunction the naïve and memory T lymphocytes which may affects the long-term effectiveness of COVID-19 vaccines. K-values of CD45 might be useful in distinguishing between severe and non-severe cases in the early infection. May be CD45 could increase the diagnostic sensitivity.


Subject(s)
Betacoronavirus/immunology , CD3 Complex/deficiency , Coronavirus Infections/immunology , Host-Pathogen Interactions/immunology , Leukocyte Common Antigens/deficiency , Pneumonia, Viral/immunology , Adolescent , Adult , Aged , Aged, 80 and over , COVID-19 , Coronavirus Infections/blood , Coronavirus Infections/diagnosis , Coronavirus Infections/virology , Early Diagnosis , Female , Humans , Male , Middle Aged , Pandemics , Pneumonia, Viral/blood , Pneumonia, Viral/diagnosis , Pneumonia, Viral/virology , Retrospective Studies , SARS-CoV-2 , Young Adult
2.
Oncogene ; 38(24): 4773-4787, 2019 06.
Article in English | MEDLINE | ID: mdl-30820040

ABSTRACT

The receptor tyrosine kinase FLT3 is expressed in myeloid and lymphoid progenitor cells. Activating mutations in FLT3 occur in 25-30% of acute myeloid leukaemia (AML) patients. Most common are internal tandem duplications of sequence (ITD) leading to constitutive FLT3-ITD kinase activity with an altered signalling quality promoting leukaemic cell transformation. Here, we observed the attenuating role of the receptor-like protein tyrosine phosphatase (RPTP) CD45/Ptprc in FLT3 signalling in vivo. Low level expression of this abundant RPTP correlates with a poor prognosis of FLT3-ITD-positive AML patients. To get a further insight into the regulatory role of Ptprc in FLT3-ITD activity in vivo, Ptprc knock-out mice were bred with FLT3-ITD knock-in mice. Inactivation of the Ptprc gene in FLT3-ITD mice resulted in a drastically shortened life span and development of severe monocytosis, a block in B-cell development and anaemia. The myeloproliferative phenotype was associated with extramedullary haematopoiesis, splenohepatomegaly and severe alterations of organ structures. The phenotypic alterations were associated with increased transforming signalling of FLT3-ITD, including activation of its downstream target STAT5. These data reveal the capacity of Ptprc for the regulation of FLT3-ITD signalling activity in vivo. In addition, histopathology and computed tomography (CT) revealed an unexpected bone phenotype; the FLT3-ITD Ptprc-/- mice, but none of the controls, showed pronounced alterations in bone morphology and, in part, apparent features of osteoporosis. In the spleen, ectopic bone formation was observed. The observed bone phenotypes suggest a previously unappreciated capacity of FLT3-ITD (and presumably FLT3) to regulate bone development/remodelling, which is under negative control of CD45/Ptprc.


Subject(s)
Bone and Bones , Leukocyte Common Antigens/genetics , Myeloproliferative Disorders/genetics , Osteoporosis/genetics , fms-Like Tyrosine Kinase 3/genetics , Animals , Bone Development/genetics , Bone Remodeling/genetics , Cell Transformation, Neoplastic , Cells, Cultured , Choristoma/genetics , Choristoma/metabolism , Embryo, Mammalian , Female , Humans , Leukemia, Myeloid, Acute/complications , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Leukocyte Common Antigens/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/pathology , Osteogenesis/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , Phenotype , Porosity , Tandem Repeat Sequences/genetics
3.
Arterioscler Thromb Vasc Biol ; 38(11): 2706-2717, 2018 11.
Article in English | MEDLINE | ID: mdl-30354229

ABSTRACT

Objective- Continuous T-cell production from thymus is essential in replenishing naïve T-cell pool and maintaining optimal T-cell functions. However, the underlying mechanisms regulating the T-cell development in thymus remains largely unknown. Approach and Results- We identified SR-BI (scavenger receptor class B type 1), an HDL (high-density lipoprotein) receptor, as a novel modulator in T-cell development. We found that SR-BI deficiency in mice led to reduced thymus size and decreased T-cell production, which was accompanied by narrowed peripheral naïve T-cell pool. Further investigation revealed that SR-BI deficiency impaired progenitor thymic homing, causing a dramatic reduction in the percentage of earliest thymic progenitors, but did not affect other downstream T-cell developmental steps inside the thymus. As a result of the impaired progenitor thymic homing, SR-BI-deficient mice displayed delayed thymic regeneration postirradiation. Using a variety of experimental approaches, we revealed that the impaired T-cell development in SR-BI-deficient mice was not caused by hematopoietic SR-BI deficiency or SR-BI deficiency-induced hypercholesterolemia, but mainly attributed to the SR-BI deficiency in adrenal glands, as adrenal-specific SR-BI-deficient mice exhibited similar defects in T-cell development and thymic regeneration with SR-BI-deficient mice. Conclusions- This study demonstrates that SR-BI deficiency impaired T-cell development and delayed thymic regeneration by affecting progenitor thymic homing in mice, elucidating a previously unrecognized link between SR-BI and adaptive immunity.


Subject(s)
Adrenal Glands/metabolism , Cell Proliferation , Lymphocyte Activation , Regeneration , Scavenger Receptors, Class B/metabolism , T-Lymphocytes/metabolism , Thymocytes/metabolism , Thymus Gland/metabolism , Adaptive Immunity , Adrenal Glands/immunology , Animals , Cells, Cultured , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Leukocyte Common Antigens/deficiency , Leukocyte Common Antigens/genetics , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Organ Size , Receptors, LDL/deficiency , Receptors, LDL/genetics , Scavenger Receptors, Class B/deficiency , Scavenger Receptors, Class B/genetics , Signal Transduction , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/immunology , Thymocytes/immunology , Thymus Gland/immunology
4.
Rinsho Ketsueki ; 58(8): 938-941, 2017.
Article in Japanese | MEDLINE | ID: mdl-28883278

ABSTRACT

A 49-year-old female was initially diagnosed with acute myeloid leukemia (AML) M4 with a CD45+CD13+CD33+CD34-HLA-DR+ immunophenotype. She underwent allogeneic bone marrow transplantation, but the disease recurred. The bone marrow was infiltrated with 87.0% blasts negative for myeloperoxidase (MPO) staining. Immunophenotyping by flow cytometry identified the presence of a CD45-negative blast population. These blasts exhibited a CD13+CD33+CD19-CD10-CD34-HLA-DR- immunophenotype. The lack of CD45 expression is often observed in B-cell acute lymphoblastic leukemia, whereas CD45-negative AML is extremely rare; only one older male with AML-M0 has been reported. In the present case, the CD45-negative blasts had an MPO-CD13+CD33+ phenotype, which is similar to AML-M0.


Subject(s)
Leukemia, Myeloid, Acute/immunology , Leukocyte Common Antigens/analysis , Fatal Outcome , Female , Flow Cytometry , Humans , Leukemia, Myeloid, Acute/pathology , Leukocyte Common Antigens/deficiency , Leukocyte Common Antigens/immunology , Middle Aged
5.
Eur J Immunol ; 47(7): 1153-1159, 2017 07.
Article in English | MEDLINE | ID: mdl-28439878

ABSTRACT

The origin of the thymic epithelium, i.e. the cortical (cTEC) and medullary (mTEC) epithelial cells, from bipotent stem cells through TEC progenitors and lineage-specific progeny still remains poorly understood. We sought to obtain an unbiased view of the incipient emergence of TEC subsets by following embryonic TEC development based on co-expression of EpCAM, CD80 and MHC class II (MHCII) on non-hematopoietic (CD45- ) thymic stromal cells in wild-type BL6 mice. Using a combination of ex vivo analysis, Re-aggregate Thymic Organ Culture (RTOC) reconstitution assays and mathematical modeling, we traced emergent lineage commitment in murine embryonic TECs. Both experimental and mathematical datasets supported the following developmental sequence: MHCII- CD80- → MHCIIlo CD80- → MHCIIhi CD80- → MHCIIhi CD80hi TECs, whereby MHCIIhi CD80- and MHCIIhi CD80hi TECs bear features of cTECs and mTECs respectively. These emergent MHCIIhi CD80- cTECs directly generate mature MHCIIhi CD80hi mTECs in vivo and in vitro, thus supporting the asynchronous model of TEC lineage commitment.


Subject(s)
Cell Differentiation , Epithelial Cells/physiology , Thymocytes/physiology , Thymus Gland/cytology , Animals , B7-1 Antigen/genetics , B7-1 Antigen/immunology , Cell Lineage , Cells, Cultured , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/immunology , Epithelial Cells/immunology , Gene Expression , Genes, MHC Class II/genetics , Genes, MHC Class II/immunology , Leukocyte Common Antigens/deficiency , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Mice , Models, Theoretical , Organ Culture Techniques , Thymus Gland/embryology , Thymus Gland/immunology
6.
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
7.
Stem Cells ; 34(11): 2733-2743, 2016 11.
Article in English | MEDLINE | ID: mdl-27339271

ABSTRACT

Bone homeostasis comprises the balance between bone-forming osteoblasts and bone-resorbing osteoclasts (OCs), with an acceleration of osteoclastic bone resorption leading to osteoporosis. OCs can be generated from bone marrow cells (BMCs) under the tightly regulated local bone environment. However, it remained difficult to identify the critical cells responsible for providing an osteoclastogenesis niche. In this study, we used a fluorescence-activated cell sorting technique to determine the cell populations important for forming an appropriate microenvironment for osteoclastogenesis and to verify the associated interactions between osteoclast precursor cells and non-OCs. We isolated and removed a small cell population specific for osteoclastogenesis (CXCR4+ CD45- ) from mouse BMCs and cultured the remaining cells with receptor activator of nuclear factor-kappa B ligand (RANKL) and macrophage-colony stimulating factor. The resulting cultures showed significantly less large osteoclast formation. Quantitative RT-PCR analysis revealed that these CXCR4+ CD45- cells expressed low levels of RANK and RANKL, but high levels of critical chemokines including stromal cell derived factor 1 (SDF-1), chemokine (C-X-C motif) ligand 7 (CXCL7), and chemokine (C-X3-C motif) ligand 1 (CX3CL1). Furthermore, an SDF-1-specific antibody strongly suppressed OC formation in RAW264.7 cells and antibodies against SDF-1, CXCL7, and CX3CL1 suppressed OC formation in BMCs. These results suggest that isolated CXCR4+ CD45- cells support an appropriate microenvironment for osteoclastogenesis with a direct effect on the cells expressing SDF-1, CXCL7, and CX3CL1 receptors. The regulation of CXCR4+ CD45- cell function might therefore inform therapeutic strategies for diseases involving loss of bone homeostasis. Stem Cells 2016;34:2733-2743.


Subject(s)
Chemokine CX3CL1/genetics , Chemokine CXCL12/genetics , Chemokines, CXC/genetics , Leukocyte Common Antigens/genetics , Osteoblasts/metabolism , Osteoclasts/metabolism , Receptors, CXCR4/genetics , Animals , Antibodies/pharmacology , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Differentiation , Chemokine CX3CL1/metabolism , Chemokine CXCL12/antagonists & inhibitors , Chemokine CXCL12/metabolism , Chemokines, CXC/metabolism , Flow Cytometry , Gene Expression Regulation , Leukocyte Common Antigens/deficiency , Macrophage Colony-Stimulating Factor/pharmacology , Male , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoclasts/cytology , Osteoclasts/drug effects , Osteogenesis/genetics , Primary Cell Culture , RANK Ligand/genetics , RANK Ligand/metabolism , RANK Ligand/pharmacology , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptors, CXCR4/metabolism , Signal Transduction , Tibia/cytology , Tibia/drug effects , Tibia/metabolism
8.
Int J Biochem Cell Biol ; 70: 1-12, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26655331

ABSTRACT

Aging is accompanied by the functional decline of cells, tissues, and organs, as well as, a striking increase in susceptibility to a wide range of diseases. Within a tissue, both differentiated cells and adult stem cells are susceptible to intrinsic and extrinsic changes while aging. Muscle derived stem cells (MDSCs) are tissue specific stem cells which have been studied well for their multipotential nature. Although there are reports relating to diminished function and regenerative capacity of aged MDSCs as compared to their young counterparts, not much has been reported relating to the concomitant gain in unipotent nature of aged MDSCs. In this study, we report an inverse correlation between aging and expression of adult/mesenchymal stem cell markers and a direct correlation between aging and myogenecity in MDSCs. Aged MDSCs were able to generate a greater number of dystrophin positive myofibres, as compared to, the young MDSCs when transplanted in muscle of dystrophic mice. Our data, therefore, suggests that aging stress adds to the decline in stem cell characteristics with a concomitant increase in unipotency, in terms of, myogenecity of MDSCs. This study, hence, also opens the possibilities of using unipotent aged MDSCs as potential candidates for transplantation in patients with muscular dystrophies.


Subject(s)
Adult Stem Cells/cytology , Aging/pathology , Antigens, CD34/genetics , Leukocyte Common Antigens/genetics , Muscle Fibers, Skeletal/cytology , Muscular Dystrophies/pathology , Adult Stem Cells/metabolism , Adult Stem Cells/transplantation , Aging/genetics , Aging/metabolism , Animals , Antigens, CD34/metabolism , Biomarkers/metabolism , Cell Differentiation , Cellular Senescence , Disease Models, Animal , Dystrophin/genetics , Dystrophin/metabolism , Flow Cytometry , Gene Expression , Leukocyte Common Antigens/deficiency , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts/cytology , Myoblasts/metabolism
9.
J Am Heart Assoc ; 4(7)2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26178404

ABSTRACT

BACKGROUND: Mammalian heart regenerative activity is lost before adulthood but increases after cardiac injury. Cardiac repair mechanisms, which involve both endogenous cardiac stem cells (CSCs) and cardiomyocyte cell-cycle reentry, are inadequate to achieve full recovery after myocardial infarction (MI). Mice deficient in S-nitrosoglutathione reductase (GSNOR(-/-)), an enzyme regulating S-nitrosothiol turnover, have preserved cardiac function after MI. Here, we tested the hypothesis that GSNOR activity modulates cardiac cell proliferation in the post-MI adult heart. METHODS AND RESULTS: GSNOR(-/-) and C57Bl6/J (wild-type [WT]) mice were subjected to sham operation (n=3 GSNOR(-/-); n=3 WT) or MI (n=41 GSNOR(-/-); n=65 WT). Compared with WT, GSNOR(-/-) mice exhibited improved survival, cardiac performance, and architecture after MI, as demonstrated by higher ejection fraction (P<0.05), lower endocardial volumes (P<0.001), and diminished scar size (P<0.05). In addition, cardiomyocytes from post-MI GSNOR(-/-) hearts exhibited faster calcium decay and sarcomeric relaxation times (P<0.001). Immunophenotypic analysis illustrated that post-MI GSNOR(-/-) hearts demonstrated enhanced neovascularization (P<0.001), c-kit(+) CSC abundance (P=0.013), and a ≈3-fold increase in proliferation of adult cardiomyocytes and c-kit(+)/CD45(-) CSCs (P<0.0001 and P=0.023, respectively) as measured by using 5-bromodeoxyuridine. CONCLUSIONS: Loss of GSNOR confers enhanced post-MI cardiac regenerative activity, characterized by enhanced turnover of cardiomyocytes and CSCs. Endogenous denitrosylases exert an inhibitory effect over cardiac repair mechanisms and therefore represents a potential novel therapeutic target.


Subject(s)
Adult Stem Cells/enzymology , Alcohol Dehydrogenase/deficiency , Cell Proliferation , Myocardial Infarction/enzymology , Myocytes, Cardiac/enzymology , Regeneration , Adult Stem Cells/pathology , Alcohol Dehydrogenase/genetics , Animals , Biomarkers/metabolism , Calcium Signaling , Cells, Cultured , Disease Models, Animal , Homozygote , Leukocyte Common Antigens/deficiency , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Neovascularization, Physiologic , Phenotype , Proto-Oncogene Proteins c-kit/metabolism , Stroke Volume , Time Factors
11.
J Vasc Res ; 51(5): 369-75, 2014.
Article in English | MEDLINE | ID: mdl-25531767

ABSTRACT

BACKGROUND/AIMS: Late-outgrowth CD45-negative endothelial colony-forming cells are implicated to be circulating endothelial progenitor cells (EPCs), as they express endothelial cell markers and can directly form blood vessels. As these cells share characteristics of other progenitor cell phenotypes, late-outgrowth EPCs were assayed for both multilineage differentiation capability and for markers of pluripotency. METHODS: Clonal single-colony late-outgrowth EPCs were derived from human cord blood and assayed both for multilineage differentiation capability in vitro and for markers of pluripotency by qPCR. RESULTS: Under osteogenic growth conditions, these EPCs expressed the osteogenic markers RUNX2, COL1A1, ALPL, and osteocalcin and demonstrated calcium deposition and bone mineralization. Endothelial colony-forming cells expressed markers associated with induced pluripotent stem cells, including SOX2, POU5F1, c-MYC, and KLF4. CONCLUSIONS: Late-outgrowth EPCs express markers associated with pluripotency and can directly express an osteogenic phenotype under bone differentiation conditions in vitro.


Subject(s)
Cell Differentiation , Endothelial Progenitor Cells/metabolism , Fetal Blood/cytology , Leukocyte Common Antigens/deficiency , Multipotent Stem Cells/metabolism , Osteogenesis , Pluripotent Stem Cells/metabolism , Biomarkers/metabolism , Cell Lineage , Cell Proliferation , Cells, Cultured , Endothelial Progenitor Cells/immunology , Humans , Kruppel-Like Factor 4 , Multipotent Stem Cells/immunology , Phenotype , Pluripotent Stem Cells/immunology , Time Factors
12.
Cell Immunol ; 288(1-2): 8-14, 2014.
Article in English | MEDLINE | ID: mdl-24556645

ABSTRACT

In this study, we analyzed intraperitoneal cells recovered from human samples and found that CD90(+)CD45(-) cells exist as a minor population but vigorously grow in culture, showing the morphological features of mesothelial cells (MC). Interestingly, the MC highly expressed arginase I and markedly suppressed T cell proliferation with the reduction of CD3 ζ chain expression in T cells stimulated by coated anti-CD3 mAb. The addition of nor-NOHA (500 µM), or L-arginine (1 mM) mostly restored the inhibitory effect of MC on T cell proliferation as well as the reduced expression of CD3 ζ chain. The expression level of CD3 ζ chain in T cells in the peritoneal cavity was significantly down-regulated from circulating T cells. These results suggest that intraperitoneal free MC have immunomodulatory functions through the control of L-arginine level, and thus may play significant roles in the pathogenesis of various diseases in the peritoneal cavity.


Subject(s)
Arginase/biosynthesis , Epithelial Cells/metabolism , Leukocyte Common Antigens/genetics , T-Lymphocytes/metabolism , Thy-1 Antigens/genetics , Antibodies, Monoclonal/pharmacology , Arginase/genetics , Arginine/analogs & derivatives , Arginine/pharmacology , CD3 Complex/genetics , CD3 Complex/metabolism , Cell Proliferation , Cells, Cultured , Coculture Techniques , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Gene Expression Regulation , Humans , Leukocyte Common Antigens/deficiency , Lymphocyte Activation/drug effects , Peritoneal Cavity/cytology , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Thy-1 Antigens/metabolism
13.
PLoS One ; 9(1): e84889, 2014.
Article in English | MEDLINE | ID: mdl-24416305

ABSTRACT

Recent studies have shown that monocytes possess pluripotent plasticity. We previously reported that monocytes could differentiate into hepatic stellate cells. Although stellate cells are also present in the pancreas, their origin remains unclear. An accumulation of enhanced green fluorescent protein (EGFP)(+)CD45(-) cells was observed in the pancreases and livers of chimeric mice, which were transplanted with a single hematopoietic stem cell isolated from EGFP-transgenic mice and treated with carbon tetrachloride (CCl4). Because the vast majority of EGFP(+)CD45(-) cells in the pancreas expressed stellate cell-associated antigens such as vimentin, desmin, glial fibrillary acidic protein, procollagen-I, and α-smooth muscle actin, they were characterized as pancreatic stellate cells (PaSCs). EGFP(+) PaSCs were also observed in CCl4-treated mice adoptively transferred with monocytes but not with other cell lineages isolated from EGFP-transgenic mice. The expression of monocyte chemoattractant protein-1 (MCP-1) and angiotensin II (Ang II) increased in the pancreas of CCl4-treated mice and their respective receptors, C-C chemokine receptor 2 (CCR2) and Ang II type 1 receptor (AT1R), were expressed on Ly6C(high) monocytes isolated from EGFP-transgenic mice. We examined the effect of an AT1R antagonist, irbesartan, which is also a CCR2 antagonist, on the migration of monocytes into the pancreas. Monocytes migrated toward MCP-1 but not Ang II in vitro. Irbesartan inhibited not only their in vitro chemotaxis but also in vivo migration of adoptively transferred monocytes from peripheral blood into the pancreas. Irbesartan treatment significantly reduced the numbers of EGFP(+)F4/80(+)CCR2(+) monocytic cells and EGFP(+) PaSCs in the pancreas of CCl4-treated chimeric mice receiving EGFP(+) bone marrow cells. A specific CCR2 antagonist RS504393 inhibited the occurrence of EGFP(+) PaSCs in injured mice. We propose that CCR2(+) monocytes migrate into the pancreas possibly via the MCP-1/CCR2 pathway and give rise to PaSCs.


Subject(s)
Cell Differentiation , Chemokine CCL2/metabolism , Chemotaxis , Monocytes/cytology , Pancreas/cytology , Pancreatic Stellate Cells/cytology , Receptors, CCR2/metabolism , Adoptive Transfer , Angiotensinogen/biosynthesis , Animals , Biphenyl Compounds/pharmacology , Carbon Tetrachloride/pharmacology , Cell Differentiation/drug effects , Chemotaxis/drug effects , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Irbesartan , Leukocyte Common Antigens/deficiency , Male , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Pancreatic Stellate Cells/drug effects , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/drug effects , Tetrazoles/pharmacology
14.
J Immunol ; 189(9): 4313-20, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23018460

ABSTRACT

Xenograft animal models using immunodeficient mice have been widely applied in medical research on various human diseases. NOD/Shi-scid-IL2rγ(null) (NOG) mice are known to show an extremely high engraftment rate of xenotransplants compared with conventional immunodeficient mice. This high engraftment rate of xenotransplants in NOG mice was substantially suppressed by the transfer of spleen cells from NOD-scid mice that were devoid of NK cells. These results indicate that cell types other than splenic NK cells present in NOD-scid mice but not in NOG mice may be involved in this suppression. To identify the cell types responsible for this effect, we transferred subpopulations of spleen cells from NOD-scid mice into NOG mice and assessed the levels of human cell engraftment after human PBMC (hPBMC) transplantation. These experiments revealed that CD11c(+)B220(+) plasmacytoid dendritic cells (pDCs) from NOD-scid mice markedly inhibited engraftment of human cells. The CD11c(+)B220(+)CD122(+) cells further fractionated from the pDCs based on the expression of CD122, which is an NK cell marker strongly inhibited during hPBMC engraftment in NOG mice. Moreover, the CD122(+) cells in the pDC fraction were morphologically distinguishable from conventional CD122(+) NK cells and showed a higher rejection efficiency. The current results suggest that CD11c(+)B220(+)CD122(+) cells play an important role in xenograft rejection, and their absence in NOG mice may be critical in supporting the successful engraftment of xenotransplants.


Subject(s)
CD11c Antigen , Graft Survival/immunology , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin-2 Receptor beta Subunit/deficiency , Leukocyte Common Antigens/deficiency , Transplantation, Heterologous/methods , Animals , CD11c Antigen/biosynthesis , CD11c Antigen/genetics , Graft Rejection/genetics , Graft Rejection/immunology , Graft Survival/genetics , Humans , Immunocompromised Host , Interleukin Receptor Common gamma Subunit/biosynthesis , Interleukin-2 Receptor beta Subunit/biosynthesis , Interleukin-2 Receptor beta Subunit/genetics , Leukocyte Common Antigens/biosynthesis , Leukocyte Common Antigens/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Spleen/immunology , Spleen/metabolism , Spleen/transplantation
15.
Proc Natl Acad Sci U S A ; 109(18): 6993-8, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22509029

ABSTRACT

Vascular smooth muscle cells (VSMC) have been suggested to arise from various developmental sources during embryogenesis, depending on the vascular bed. However, evidence also points to a common subpopulation of vascular progenitor cells predisposed to VSMC fate in the embryo. In the present study, we use binary transgenic reporter mice to identify a Tie1(+)CD31(dim)vascular endothelial (VE)-cadherin(-)CD45(-) precursor that gives rise to VSMC in vivo in all vascular beds examined. This precursor does not represent a mature endothelial cell, because a VE-cadherin promoter-driven reporter shows no expression in VSMC during murine development. Blockade of Notch signaling in the Tie1(+) precursor cell, but not the VE-cadherin(+) endothelial cell, decreases VSMC investment of developing arteries, leading to localized hemorrhage in the embryo at the time of vascular maturation. However, Notch signaling is not required in the Tie1(+) precursor after establishment of a stable artery. Thus, Notch activity is required in the differentiation of a Tie1(+) local precursor to VSMC in a spatiotemporal fashion across all vascular beds.


Subject(s)
Cell Differentiation/physiology , Myoblasts, Smooth Muscle/cytology , Myoblasts, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic , Receptors, Notch/metabolism , Animals , Antigens, CD/genetics , Arteries/embryology , Arteries/growth & development , Arteries/metabolism , Base Sequence , Cadherins/deficiency , Cadherins/genetics , Cell Differentiation/genetics , DNA Primers/genetics , Female , Leukocyte Common Antigens/deficiency , Leukocyte Common Antigens/genetics , Mice , Mice, Transgenic , Neovascularization, Physiologic/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pregnancy , Receptor, TIE-1/metabolism , Receptors, Notch/antagonists & inhibitors , Signal Transduction
16.
J Vis Exp ; (56): e3159, 2011 Oct 26.
Article in English | MEDLINE | ID: mdl-22064472

ABSTRACT

Tissue resident mesenchymal stem cells (MSC) are important regulators of tissue repair or regeneration, fibrosis, inflammation, angiogenesis and tumor formation. Taken together these studies suggest that resident lung MSC play a role during pulmonary tissue homeostasis, injury and repair during diseases such as pulmonary fibrosis (PF) and arterial hypertension (PAH). Here we describe a technology to define a population of resident lung MSC. The definition of this population in vivo pulmonary tissue using a define set of markers facilitates the repeated isolation of a well-characterized stem cell population by flow cytometry and the study of a specific cell type and function.


Subject(s)
Benzimidazoles/chemistry , Leukocyte Common Antigens/deficiency , Lung/cytology , Mesenchymal Stem Cells/cytology , Animals , Cell Separation/methods , Cytological Techniques/methods , Flow Cytometry/methods , Leukocyte Common Antigens/biosynthesis , Lung/immunology , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred C57BL
17.
J Biol Chem ; 286(25): 22101-12, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21543337

ABSTRACT

CD148 is a receptor-like protein-tyrosine phosphatase known to inhibit transduction of mitogenic signals in non-hematopoietic cells. Similarly, in the hematopoietic lineage, CD148 inhibited signal transduction downstream of T cell receptor. However, it also augmented immunoreceptor signaling in B cells and macrophages via dephosphorylating C-terminal tyrosine of Src family kinases (SFK). Accordingly, endogenous CD148 compensated for the loss of the main SFK activator CD45 in murine B cells and macrophages but not in T cells. Hypothetical explanations for the difference between T cells and other leukocyte lineages include the inability of CD148 to dephosphorylate a specific set of SFKs involved in T cell activation or the lack of CD148 expression during critical stages of T cell development. Here we describe striking differences in CD148 expression between human and murine thymocyte subsets, the only unifying feature being the absence of CD148 during the positive selection when the major developmental block occurs under CD45 deficiency. Moreover, we demonstrate that similar to CD45, CD148 has both activating and inhibitory effects on the SFKs involved in TCR signaling. However, in the absence of CD45, activating effects prevail, resulting in functional complementation of CD45 deficiency in human T cell lines. Importantly, this is independent of the tyrosines in the CD148 C-terminal tail, contradicting the recently proposed phosphotyrosine displacement model as a mechanism of SFK activation by CD148. Collectively, our data suggest that differential effects of CD148 in T cells and other leukocyte subsets cannot be explained by the CD148 inability to activate T cell SFKs but rather by its dual inhibitory/activatory function and specific expression pattern.


Subject(s)
Receptors, Antigen, T-Cell/metabolism , Signal Transduction , src-Family Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Enzyme Activation , Gene Expression Regulation, Enzymologic , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/metabolism , Humans , Jurkat Cells , Leukocyte Common Antigens/deficiency , Membrane Proteins/metabolism , Mice , Phospholipase C gamma/metabolism , Phosphorylation , Protein Structure, Tertiary , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , T-Lymphocytes/metabolism , Thymus Gland/cytology , Tyrosine/metabolism , src-Family Kinases/chemistry
18.
Nature ; 472(7344): 471-5, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21525931

ABSTRACT

Innate immune cells must be able to distinguish between direct binding to microbes and detection of components shed from the surface of microbes located at a distance. Dectin-1 (also known as CLEC7A) is a pattern-recognition receptor expressed by myeloid phagocytes (macrophages, dendritic cells and neutrophils) that detects ß-glucans in fungal cell walls and triggers direct cellular antimicrobial activity, including phagocytosis and production of reactive oxygen species (ROS). In contrast to inflammatory responses stimulated upon detection of soluble ligands by other pattern-recognition receptors, such as Toll-like receptors (TLRs), these responses are only useful when a cell comes into direct contact with a microbe and must not be spuriously activated by soluble stimuli. In this study we show that, despite its ability to bind both soluble and particulate ß-glucan polymers, Dectin-1 signalling is only activated by particulate ß-glucans, which cluster the receptor in synapse-like structures from which regulatory tyrosine phosphatases CD45 and CD148 (also known as PTPRC and PTPRJ, respectively) are excluded (Supplementary Fig. 1). The 'phagocytic synapse' now provides a model mechanism by which innate immune receptors can distinguish direct microbial contact from detection of microbes at a distance, thereby initiating direct cellular antimicrobial responses only when they are required.


Subject(s)
Immunity, Innate/immunology , Immunological Synapses/immunology , Membrane Proteins/immunology , Models, Immunological , Nerve Tissue Proteins/immunology , Phagocytosis/immunology , Animals , Cell Wall/chemistry , Cell Wall/immunology , Cells, Cultured , Humans , Lectins, C-Type , Leukocyte Common Antigens/deficiency , Leukocyte Common Antigens/metabolism , Macrophages/immunology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Reactive Oxygen Species/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/immunology , Signal Transduction/immunology , Solubility , beta-Glucans/chemistry , beta-Glucans/immunology
19.
Blood ; 117(11): 3087-95, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21245479

ABSTRACT

The protein tyrosine phosphatase CD45 is an important regulator of Src-family kinase activity. We found that in the absence of CD45, natural killer (NK) cells are defective in protecting the host from mouse cytomegalovirus infection. We show that although CD45 is necessary for all immunoreceptor tyrosine-based activation motif (ITAM)-specific NK-cell functions and processes such as degranulation, cytokine production, and expansion during viral infection, the impact of CD45 deficiency on ITAM signaling differs depending on the downstream function. CD45-deficient NK cells are normal in their response to inflammatory cytokines when administered ex vivo and in the context of viral infection. Syk and ζ chain-associated protein kinase 70 (Zap70) are thought to play redundant roles in transmitting ITAM signals in NK cells. We show that Syk, but not Zap70, controls the remaining CD45-independent, ITAM-specific NK-cell functions, demonstrating a functional difference between these 2 Syk-kinase family members in primary NK cells.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Killer Cells, Natural/enzymology , Killer Cells, Natural/immunology , Leukocyte Common Antigens/metabolism , Protein-Tyrosine Kinases/metabolism , Amino Acid Motifs , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Degranulation/immunology , Cell Proliferation , Herpesviridae Infections/immunology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/virology , Interferon-gamma/biosynthesis , Intracellular Signaling Peptides and Proteins/deficiency , Killer Cells, Natural/physiology , Leukocyte Common Antigens/deficiency , Mice , Muromegalovirus/physiology , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Protein-Tyrosine Kinases/deficiency , Receptors, Virus/metabolism , Syk Kinase , ZAP-70 Protein-Tyrosine Kinase/metabolism
20.
Am J Physiol Gastrointest Liver Physiol ; 300(3): G409-17, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21183658

ABSTRACT

Intestinal stem cells (ISCs) have been studied for more than three decades; however, their isolation has remained a challenge. We hypothesized that, just as for stem cells of other tissues, one or more membrane markers would allow positive selection of ISCs by antibody-based sorting. To explore this hypothesis, microarray data of putative ISC fractions generated by side population sorting and laser capture microdissection were subjected to bioinformatic analysis to identify common membrane antigens. The microarray comparison suggested CD24 as a candidate surface marker, and immunohistochemistry showed expression of CD24 in epithelial cells of crypt bases. Flow cytometry of jejunal epithelial preparations revealed a CD24(+) CD45(-) fraction comprising ∼1% of the cells. Analysis with epithelial cell adhesion molecule and CD31 confirmed that the cell preparations were epithelial and without endothelial contamination. Cycling cells identified by prior injection with 5-ethynyl-2'-deoxyuridine were found predominantly in the CD24(lo) subfraction. Transcript analysis by real-time RT-PCR showed this subfraction to be enriched in the ISC markers leucine-rich-repeat-containing G-protein-coupled receptor 5 (40-fold) and Bmi1 (5-fold), but also enriched in lysozyme (10-fold). Flow cytometry with anti-lysozyme antibodies demonstrated that Paneth cells comprise ∼30% of the CD24(lo) subfraction. Additional flow analyses with leucine-rich-repeat-containing G-protein-coupled receptor 5-enhanced green fluorescent protein (EGFP) epithelium demonstrated colocalization of EGFP(hi) and CD24(lo). In contrast, CD24 cells were negative for the quiescent ISC marker doublecortin and CaM kinase-like-1. Culture of CD24(lo) cells in Matrigel generated organoid structures, which included all four epithelial lineages, thus giving functional evidence for the presence of ISCs. We conclude that the CD24(lo) fraction of jejunal epithelium is highly enriched with cycling ISCs. This isolation method should be useful to many investigators in the field to advance both the basic understanding of ISC biology and the therapeutic applications of ISCs.


Subject(s)
CD24 Antigen/metabolism , Cell Separation/methods , Epithelial Cells/immunology , Flow Cytometry , Jejunum/immunology , Paneth Cells/immunology , Stem Cells/immunology , Animals , Biomarkers/metabolism , CD24 Antigen/genetics , Cell Adhesion Molecules/metabolism , Cell Proliferation , Cells, Cultured , Doublecortin-Like Kinases , Epithelial Cells/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation , Immunohistochemistry , Jejunum/cytology , Jejunum/metabolism , Leukocyte Common Antigens/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Paneth Cells/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Polycomb Repressive Complex 1 , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...