Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
JCI Insight ; 52019 03 26.
Article in English | MEDLINE | ID: mdl-30912768

ABSTRACT

Tregs require IL-2 signaling for signal transducer and activator of transcription 5 (STAT5)-mediated induction of Foxp3. While phosphatase 2A (PP2A) is a negative regulator of IL-2 production in effector T cells and Tregs do not produce IL-2, it is not known whether PP2A controls IL-2 signaling in Tregs. To address the role of PP2A in IL-2 signaling in Tregs we studied mice engineered to lack PP2A in all Foxp3-expressing cells. We report that PP2A is required to enable Foxp3 expression and to maintain sufficient numbers of Tregs in the thymus. We show for the first time that PP2A prevents the selective loss of surface IL-2Rß and preserves IL-2R signaling potency in Tregs. The loss of IL-2Rß in thymus- and spleen-derived Tregs that lack PP2A is due to increased sheddase activity. Pan-sheddase or selective A disintegrin and metalloproteinase 10 (ADAM10) inhibition, like forced expression of IL-2Rß in PP2A-deficient Tregs restored IL-2Rß expression and signaling. Thus, PP2A restrains the sheddase activity of ADAM10 in Treg cells to prevent the cleavage of IL-2Rß from the cell surface to enable competent IL-2R signaling which is essential for Tregs development and homeostasis.


Subject(s)
Interleukin-2/metabolism , Protein Phosphatase 2/metabolism , Receptors, Interleukin-2/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases , Animals , Autoimmune Diseases/metabolism , Autoimmunity , CD4-Positive T-Lymphocytes/immunology , Disintegrins/metabolism , Forkhead Transcription Factors/metabolism , Homeostasis , Membrane Proteins , Mice , Mice, Knockout , Protein Phosphatase 2/genetics , Receptors, Interleukin-2/deficiency , STAT5 Transcription Factor/metabolism , Spleen/pathology , Thymus Gland/pathology
2.
J Pharmacol Exp Ther ; 360(1): 174-191, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27789682

ABSTRACT

The humanized liver mouse model is being exploited increasingly for human drug metabolism studies. However, its model stability, intercommunication between human hepatocytes and mouse nonparenchymal cells in liver and murine intestine, and changes in extrahepatic transporter and enzyme expressions have not been investigated. We examined these issues in FRGN [fumarylacetoacetate hydrolase (Fah-/-), recombination activating gene 2 (Rag2-/-), and interleukin 2 receptor subunit gamma (IL-2rg -/-) triple knockout] on nonobese diabetic (NOD) background] and chimeric mice: mFRGN and hFRGN (repopulated with mouse or human hepatocytes, respectively). hFRGN mice showed markedly higher levels of liver cholesterol, biliary bilirubin, and bile acids (liver, bile, and plasma; mainly human forms, but also murine bile acids) but lower transforming growth factor beta receptor 2 (TGFBR2) mRNA expression levels (10%) in human hepatocytes and other proliferative markers in mouse nonparenchymal cells (Tgf-ß1) and cholangiocytes [plasma membrane-bound, G protein-coupled receptor for bile acids (Tgr5)], suggestive of irregular regeneration processes in hFRGN livers. Changes in gene expression in murine intestine, kidney, and brain of hFRGN mice, in particular, induction of intestinal farnesoid X receptor (Fxr) genes: fibroblast growth factor 15 (Fgf15), mouse ileal bile acid binding protein (Ibabp), small heterodimer partner (Shp), and the organic solute transporter alpha (Ostα), were observed. Proteomics revealed persistence of remnant murine proteins (cyotchrome P450 7α-hydroxylase (Cyp7a1) and other enzymes and transporters) in hFRGN livers and suggest the likelihood of mouse activity. When compared with normal human liver tissue, hFRGN livers showed lower SHP mRNA and higher CYP7A1 (300%) protein expression, consequences of tß- and tα-muricholic acid-mediated inhibition of the FXR-SHP cascade and miscommunication between intestinal Fgf15 and human liver fibroblast growth factor receptor 4 (FGFR4), as confirmed by the unchanged hepatic pERK/total ERK ratio. Dysregulation of hepatocyte proliferation and bile acid homeostasis in hFRGN livers led to hepatotoxicity, gallbladder distension, liver deformity, and other extrahepatic changes, making questionable the use of the preparation for drug metabolism studies.


Subject(s)
Bile Acids and Salts/metabolism , Homeostasis , Intestines/cytology , Liver/cytology , Liver/metabolism , Signal Transduction , Adolescent , Adult , Animals , Bile Acids and Salts/blood , Child , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation , Gene Knockout Techniques , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Hydrolases/deficiency , Hydrolases/genetics , Male , Mice , Receptors, Interleukin-2/deficiency , Receptors, Interleukin-2/genetics
3.
Br J Cancer ; 115(8): 920-928, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27560553

ABSTRACT

BACKGROUND: Renal cell carcinoma (RCC) patients treated with tyrosine kinase inhibitors (TKI) typically respond initially, but usually develop resistance to therapy. We utilised transcriptome analysis to identify gene expression changes during development of sunitinib resistance in a RCC patient-derived xenograft (PDX) model. METHODS: RCC tumours were harvested during pre-treatment, response and escape phases. Direct anti-proliferative effects of sunitinib plus MEK inhibitor were assessed. Activation status (phosphorylation) of MEK1/2 and ERK1/2 was determined, myeloid-derived suppressor cells (MDSC) sub-fractions were quantitated and G-CSF was measured by ELISA. RESULTS: During the response phase, tumours exhibited 91% reduction in volume, characterised by decreased expression of cell survival genes. After 4-week treatment, tumours developed resistance to sunitinib, associated with increased expression of pro-angiogenic and cell survival genes. During tumour escape, cellular movement, inflammatory response and immune cell trafficking genes were induced, along with intra-tumoural accumulation of MDSC. In this PDX model, either continuous treatment with sunitinib plus MEK inhibitor PD-0325901, or switching from sunitinib to PD-0325901 was effective. The combination of PD-0325901 with TKI suppressed intra-tumoural phospho-MEK1/2, phospho-ERK1/2 and MDSC. CONCLUSIONS: Continuous treatment with sunitinib alone did not maintain anti-tumour response; addition of MEK inhibitor abrogated resistance, leading to improved anti-tumour efficacy.


Subject(s)
Benzamides/therapeutic use , Carcinoma, Renal Cell/drug therapy , Diphenylamine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Indoles/pharmacology , Kidney Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Adult , Animals , Benzamides/pharmacology , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/immunology , Cell Line, Tumor , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/therapeutic use , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Kidney Neoplasms/immunology , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , Protein Processing, Post-Translational/drug effects , Pyrroles/therapeutic use , Receptors, Interleukin-2/deficiency , Sunitinib , Tumor Burden/drug effects , Tumor Escape/drug effects , Xenograft Model Antitumor Assays
4.
Cell Rep ; 14(10): 2281-8, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26947068

ABSTRACT

Forced overexpression and/or downregulation of proteins regulating epithelial-to-mesenchymal transition (EMT) has been reported to alter metastasis by changing migration and stem cell capacity of tumor cells. However, these manipulations artificially keep cells in fixed states, while in vivo cells may adapt transient and reversible states. Here, we have tested the existence and role of epithelial-mesenchymal plasticity in metastasis of mammary tumors without artificially modifying EMT regulators. In these tumors, we found by intravital microscopy that the motile tumor cells have undergone EMT, while their epithelial counterparts were not migratory. Moreover, we found that epithelial-mesenchymal plasticity renders any EMT-induced stemness differences, as reported previously, irrelevant for metastatic outgrowth, because mesenchymal cells that arrive at secondary sites convert to the epithelial state within one or two divisions, thereby obtaining the same stem cell potential as their arrived epithelial counterparts. We conclude that epithelial-mesenchymal plasticity supports migration but additionally eliminates stemness-enhanced metastatic outgrowth differences.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal/pathology , Epithelial-Mesenchymal Transition , Neoplastic Stem Cells/metabolism , Animals , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Breast Neoplasms/etiology , Breast Neoplasms/metabolism , Cadherins/antagonists & inhibitors , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Ductal/etiology , Carcinoma, Ductal/metabolism , Disease Models, Animal , Female , Flow Cytometry , Humans , Immunohistochemistry , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice , Mice, Knockout , Mice, SCID , Neoplastic Stem Cells/cytology , RNA Interference , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-2/deficiency , Receptors, Interleukin-2/genetics
5.
Gastroenterology ; 149(3): 728-40.e15, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26028580

ABSTRACT

BACKGROUND & AIMS: The ratio of liver size to body weight (hepatostat) is tightly controlled, but little is known about how the physiologic functions of the liver help determine its size. Livers of mice repopulated with human hepatocytes (humanized livers) grow to larger than normal; the human hepatocytes do not recognize the fibroblast growth factor (FGF)-15 produced by mouse intestine. This results in up-regulation of bile acid synthesis in the human hepatocytes and enlargement of the bile acid pool. We investigated whether abnormal bile acid signaling affects the hepatostat in mice. METHODS: We crossed Fah(-/-), Rag2(-/-), Il2r(-/-) mice with nonobese diabetic mice to create FRGN mice, whose livers can be fully repopulated with human hepatocytes. We inserted the gene for human FGF19 (ortholog to mouse Fgf15), including regulatory sequences, into the FRGN mice to create FRGN19(+) mice. Livers of FRGN19(+) mice and their FRGN littermates were fully repopulated with human hepatocytes. Liver tissues were collected and bile acid pool sizes and RNA sequences were analyzed and compared with those of mice without humanized livers (controls). RESULTS: Livers were larger in FRGN mice with humanized livers (13% of body weight), compared with control FRGN mice; they also had much larger bile acid pools and aberrant bile acid signaling. Livers from FRGN19(+) normalized to 7.8% of body weight, and their bile acid pool and signaling more closely resembled that of control FRGN19(+) mice. RNA sequence analysis showed activation of the Hippo pathway, and immunohistochemical and transcription analyses revealed increased hepatocyte proliferation, but not apoptosis, in the enlarged humanized livers of FRGN mice. Cell sorting experiments showed that although healthy human liver does not produce FGF19, nonparenchymal cells from cholestatic livers produce FGF19. CONCLUSIONS: In mice with humanized livers, expression of an FGF19 transgene corrects bile acid signaling defects, resulting in normalization of bile acid synthesis, the bile acid pool, and liver size. These findings indicate that liver size is, in part, regulated by the size of the bile acid pool that the liver must circulate.


Subject(s)
Cell Proliferation , Fibroblast Growth Factors/biosynthesis , Fibroblast Growth Factors/metabolism , Hepatocytes/transplantation , Liver/surgery , Signal Transduction , Animals , Bile Acids and Salts/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Fibroblast Growth Factors/genetics , Gene Expression Regulation , Hepatocytes/metabolism , Heterografts , Humans , Hydrolases/deficiency , Hydrolases/genetics , Intestinal Mucosa/metabolism , Liver/metabolism , Mice, Knockout , Mice, Transgenic , Organ Size , Receptors, Interleukin-2/deficiency , Receptors, Interleukin-2/genetics , Time Factors , Transcription, Genetic
6.
Stem Cells ; 33(6): 1975-84, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25753525

ABSTRACT

Simple efforts are needed to enhance cord blood (CB) transplantation. We hypothesized that short-term exposure of CD34(+) CB cells to 39.5°C would enhance their response to stromal-derived factor-1 (SDF-1), by increasing lipid raft aggregation and CXCR4 expression, thus leading to enhanced engraftment. Mild hyperthermia (39.5°C) significantly increased the percent of CD34(+) CB that migrated toward SDF-1. This was associated with increased expression of CXCR4 on the cells. Mechanistically, mild heating increased the percent of CD34(+) cells with aggregated lipid rafts and enhanced colocalization of CXCR4 within lipid raft domains. Using methyl-ß-cyclodextrin (MßCD), an agent that blocks lipid raft aggregation, it was determined that this enhancement in chemotaxis was dependent upon lipid raft aggregation. Colocalization of Rac1, a GTPase crucial for cell migration and adhesion, with CXCR4 to the lipid raft was essential for the effects of heat on chemotaxis, as determined with an inhibitor of Rac1 activation, NSC23766. Application-wise, mild heat treatment significantly increased the percent chimerism as well as homing and engraftment of CD34(+) CB cells in sublethally irradiated non-obese diabetic severe combined immunodeficiency IL-2 receptor gamma chain d (NSG) mice. Mild heating may be a simple and inexpensive means to enhance engraftment following CB transplantation in patients.


Subject(s)
Cell Movement , Chemokine CXCL12/metabolism , Fetal Blood/cytology , Animals , Antigens, CD34/immunology , Blood Cells/cytology , Bone Marrow/metabolism , Female , Hematopoietic Stem Cell Transplantation/methods , Hot Temperature , Humans , Receptors, CXCR4/metabolism , Receptors, Interleukin-2/deficiency
7.
Exp Anim ; 63(3): 321-30, 2014.
Article in English | MEDLINE | ID: mdl-25077762

ABSTRACT

We have developed NOD-Rag2(null) IL-2Rγ(null) (NR2G) mice similar to NOD-scidIL-2Rγ(null) (NOG) mice that are known as an excellent host to generate humanized mice. To evaluate the usefulness of NR2G mice as a host for humanized mice, the engraftment rates and differentiation of human cells after human hematopoietic stem cell (HSC) transplantation were compared among NR2G, NOG, and NOD-scid mice. For this purpose, the appropriate irradiation doses to expand the niche for human stem cells in the bone marrow were first determined. As a result, 8 and 2.5 Gy in adult, and 4 and 1 Gy in newborn NR2G and NOG mice, respectively, were found to be appropriate. Next, 5 × 10(4) human umbilical cord blood CD34(+) cells were intravenously inoculated into irradiated adult or newborn of the immunodeficient mice. These HSC transplantation experiments demonstrated that both NR2G and NOG mice showed high engraftment rates compared with NOD-scid mice, although NOG mice showed a slightly higher engraftment rate than that for NR2G mice. However, no difference was found in the human cell populations differentiated from HSCs between NR2G and NOG mice. The HSC transplantation experiments to adults and newborns of two immunodeficient mice also revealed that the HSC transplantation into newborn mice resulted in higher engraftment rate than those into adults. These results showed that NR2G mice could be used as an alternative host to NOG mice to generate humanized mice.


Subject(s)
Disease Models, Animal , Animals , Animals, Newborn , Antigens, CD34 , DNA-Binding Proteins/deficiency , Fetal Blood/transplantation , Graft Survival , Hematopoietic Stem Cell Transplantation , Humans , Mice, Inbred NOD , Mice, SCID , Radiation Dosage , Receptors, Interleukin-2/deficiency , Whole-Body Irradiation
8.
J Immunol ; 192(3): 1044-54, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24379121

ABSTRACT

The peripheral B cell compartment is maintained by homeostatic proliferation and through replenishment by bone marrow precursors. Because hematopoietic stem cells cycle at a slow rate, replenishment must involve replication of precursor B cells. To study proliferation of early human B cell progenitors, we established a feeder cell-free in vitro system allowing the development of B cells from CD34(+) hematopoietic stem cells up to the stage of immature IgM(+) B cells. We found that pro-B and pre-B cells generated in vitro can proliferate autonomously and persist up to 7 wk in culture in the absence of signals induced by exogenously added cytokines. Nevertheless, addition of IL-7 enhanced pre-B cell expansion and inhibited maturation into IgM(+) B cells. The B cell precursor subsets replicating in vitro were highly similar to the bone marrow B cell precursors cycling in vivo. The autonomous proliferation of B cell precursor subsets in vitro and their long-term persistence implies that proliferation during pro-B and pre-B cell stages plays an important role in the homeostasis of the peripheral B cell compartment. Our in vitro culture can be used to study defects in B cell development or in reconstitution of the B cell pool after depletion and chemotherapy.


Subject(s)
B-Lymphocytes/cytology , Cell Culture Techniques/methods , Hematopoietic Stem Cells/cytology , Adult , Animals , Bone Marrow , Cell Division , Cell Lineage , Cells, Cultured , Coculture Techniques , DNA-Binding Proteins/deficiency , Fetal Blood/cytology , Graft Survival , Hematopoietic Stem Cells/drug effects , Heterografts , Homeostasis , Humans , Immunoglobulin M/biosynthesis , Immunophenotyping , Interleukin-7/pharmacology , Lymphopoiesis/drug effects , Mice , Radiation Chimera , Receptors, Interleukin-2/deficiency , Time Factors , Young Adult
9.
PLoS One ; 8(11): e79939, 2013.
Article in English | MEDLINE | ID: mdl-24223204

ABSTRACT

BACKGROUND: We systematically analyzed multiple myeloma (MM) cell lines and patient bone marrow cells for their engraftment capacity in immunodeficient mice and validated the response of the resulting xenografts to antimyeloma agents. DESIGN AND METHODS: Using flow cytometry and near infrared fluorescence in-vivo-imaging, growth kinetics of MM cell lines L363 and RPMI8226 and patient bone marrow cells were investigated with use of a murine subcutaneous bone implant, intratibial and intravenous approach in NOD/SCID, NOD/SCID treated with CD122 antibody and NOD/SCID IL-2Rγ(null) mice (NSG). RESULTS: Myeloma growth was significantly increased in the absence of natural killer cell activity (NSG or αCD122-treated NOD/SCID). Comparison of NSG and αCD122-treated NOD/SCID revealed enhanced growth kinetics in the former, especially with respect to metastatic tumor sites which were exclusively observed therein. In NSG, MM cells were more tumorigenic when injected intratibially than intravenously. In NOD/SCID in contrast, the use of juvenile long bone implants was superior to intratibial or intravenous cancer cell injection. Using the intratibial NSG model, mice developed typical disease symptoms exclusively when implanted with human MM cell lines or patient-derived bone marrow cells, but not with healthy bone marrow cells nor in mock-injected animals. Bortezomib and dexamethasone delayed myeloma progression in L363- as well as patient-derived MM cell bearing NSG. Antitumor activity could be quantified via flow cytometry and in vivo imaging analyses. CONCLUSIONS: Our results suggest that the intratibial NSG MM model mimics the clinical situation of the disseminated disease and serves as a valuable tool in the development of novel anticancer strategies.


Subject(s)
Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Receptors, Interleukin-2/deficiency , Animals , Boronic Acids/therapeutic use , Bortezomib , Cell Line, Tumor , Dexamethasone/therapeutic use , Disease Models, Animal , Flow Cytometry , Humans , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/drug therapy , Pyrazines/therapeutic use , Receptors, Interleukin-2/genetics
10.
PLoS One ; 8(8): e71270, 2013.
Article in English | MEDLINE | ID: mdl-23967178

ABSTRACT

We compare for the first time, the metastatic aggressiveness of the parental MDA-MB-231 breast cancer cell line and two luciferase-tagged in vivo-derived and selected pro-metastatic variants (LM2-4/luc⁺ and 164/8-1B/luc⁺ in SCID, NOD-SCID and NOD-SCID-IL-2Rγ(null) (NSG) mice following orthotopic implantation and primary tumour resection. The variants are known to be more aggressively metastatic in SCID mice, compared to the parental line which has limited spontaneous metastatic competence in these mice. When 2×106 cells were injected into the mammary fat pad, the growth of the resultant primary tumours was identical for the various cell lines in the three strains of mice. However, metastatic spread of all three cell lines, including the MDA-MB-231 parental cell line, was strikingly more aggressive in the highly immunocompromised NSG mice compared to both NOD-SCID and SCID mice, resulting in extensive multi-organ metastases and a significant reduction in overall survival. While these studies were facilitated by monitoring post-surgical spontaneous metastases using whole body bioluminescence imaging, we observed that the luciferase-tagged parental line showed altered growth and diminished metastatic properties compared to its untagged counterpart. Our results are the first to show that host immunity can have a profound impact on the spread of spontaneous visceral metastases and survival following resection of a primary tumour in circumstances where the growth of primary tumours is not similarly affected; as such they highlight the importance of immunity in the metastatic process, and by extension, suggest certain therapeutic strategies that may have a significant impact on reducing metastasis.


Subject(s)
Gene Deletion , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/surgery , Receptors, Interleukin-2/deficiency , Receptors, Interleukin-2/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Female , Humans , Luciferases, Firefly/genetics , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Grading , Neoplasm Metastasis , Survival Analysis
11.
J Immunol ; 190(4): 1567-75, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23315074

ABSTRACT

CD4(+) Foxp3(+) regulatory T cells (Tregs) are an independent cell lineage, and their developmental progression during thymic development depends on IL-2R signaling. However, the role of IL-2R signaling during thymic Treg development remains only partially understood. The current study assessed the contribution of IL-2 to the expansion and functional programming of developing Tregs. In the absence of IL-2Rß signaling, predominantly CD4(+) CD25(-) Foxp3(lo) T cells were found, and these cells exhibited somewhat lower expression of the proliferative marker Ki67. These immature Tregs, which represent products of failed development, were also found in normal mice and were characterized by markedly lower expression of several Treg functional molecules. Therefore, IL-2R is required for the progression, functional programming, and expansion of Tregs during thymic development. An IL-2R-signaling mutant that lowers STAT5 activation readily supported Treg functional programming, but Treg proliferation remained somewhat impaired. The requirement for IL-2 during thymic Treg expansion was best illustrated in mixed chimeras where the Tregs with mutant IL-2Rs were forced to compete with wild-type Tregs during their development. Tregs with impaired IL-2R signaling were more prevalent in the thymus than spleen in these competitive experiments. The general effectiveness of mutant IL-2Rs to support thymic Treg development is partially accounted for by a heightened capacity of thymic Tregs to respond to IL-2. Overall, our data support a model in which limiting IL-2R signaling is amplified by thymic Tregs to readily support their development and functional programming, whereas these same conditions are not sufficient to support peripheral Treg homeostasis.


Subject(s)
Cell Differentiation/immunology , Receptors, Interleukin-2/physiology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Animals , Cell Differentiation/genetics , Forkhead Transcription Factors/biosynthesis , Genes, Reporter , Interleukin-2 Receptor beta Subunit/deficiency , Interleukin-2 Receptor beta Subunit/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Interleukin-2/deficiency , Signal Transduction/genetics , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/embryology , Thymus Gland/growth & development
12.
Gastroenterology ; 142(4): 886-896.e9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22240480

ABSTRACT

BACKGROUND & AIMS: MicroRNAs (miRNAs) can promote or inhibit tumor growth and are therefore being developed as targets for cancer therapies. They are diverse not only in the messenger RNAs (mRNA) they target, but in their production; the same hairpin RNA structure can generate mature products from each strand, termed 5p and 3p, that can bind different mRNAs. We analyzed the expression, functions, and mechanisms of miR-28-5p and miR-28-3p in colorectal cancer (CRC) cells. METHODS: We measured levels of miR-28-5p and miR-28-3p expression in 108 CRC and 49 normal colorectal samples (47 paired) by reverse transcription, quantitative real-time polymerase chain reaction. The roles of miR-28 in CRC development were studied using cultured HCT116, RKO, and SW480 cells and tumor xenograft analyses in immunodeficient mice; their mRNA targets were also investigated. RESULTS: miR-28-5p and miR-28-3p were down-regulated in CRC samples compared with normal colon samples. Overexpression of miRNAs in CRC cells had different effects and the miRNAs interacted with different mRNAs: miR-28-5p altered expression of CCND1 and HOXB3, whereas miR-28-3p bound NM23-H1. Overexpression of miR-28-5p reduced CRC cell proliferation, migration, and invasion in vitro, whereas miR-28-3p increased CRC cell migration and invasion in vitro. CRC cells overexpressing miR-28 developed tumors more slowly in mice compared with control cells, but miR-28 promoted tumor metastasis in mice. CONCLUSION: miR-28-5p and miR-28-3p are transcribed from the same RNA hairpin and are down-regulated in CRC cells. Overexpression of each has different effects on CRC cell proliferation and migration. Such information has a direct application for the design of miRNA gene therapy trials.


Subject(s)
Colorectal Neoplasms/therapy , Genetic Therapy , MicroRNAs/metabolism , Animals , Apoptosis , Case-Control Studies , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cyclin D1/genetics , Cyclin D1/metabolism , G1 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , HCT116 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , NM23 Nucleoside Diphosphate Kinases/genetics , NM23 Nucleoside Diphosphate Kinases/metabolism , Neoplasm Invasiveness , RNA Interference , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-2/deficiency , Receptors, Interleukin-2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection , Tumor Burden , Xenograft Model Antitumor Assays
13.
Proc Natl Acad Sci U S A ; 108(32): 13224-9, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21788504

ABSTRACT

The homeostatic control mechanisms regulating human leukocyte numbers are poorly understood. Here, we assessed the role of phagocytes in this process using human immune system (HIS) BALB/c Rag2(-/-)IL-2Rγc(-/-) mice in which human leukocytes are generated from transplanted hematopoietic progenitor cells. Interactions between signal regulatory protein alpha (SIRPα; expressed on phagocytes) and CD47 (expressed on hematopoietic cells) negatively regulate phagocyte activity of macrophages and other phagocytic cells. We previously showed that B cells develop and survive robustly in HIS mice, whereas T and natural killer (NK) cells survive poorly. Because human CD47 does not interact with BALB/c mouse SIRPα, we introduced functional CD47/SIRPα interactions in HIS mice by transducing mouse CD47 into human progenitor cells. Here, we show that this procedure resulted in a dramatic and selective improvement of progenitor cell engraftment and human T- and NK-cell homeostasis in HIS mouse peripheral lymphoid organs. The amount of engrafted human B cells also increased but much less than that of T and NK cells, and total plasma IgM and IgG concentrations increased 68- and 35-fold, respectively. Whereas T cells exhibit an activated/memory phenotype in the absence of functional CD47/SIRPα interactions, human T cells accumulated as CD4(+) or CD8(+) single-positive, naive, resting T cells in the presence of functional CD47/SIRPα interactions. Thus, in addition to signals mediated by T cell receptor (TCR)/MHC and/or IL/IL receptor interactions, sensing of cell surface CD47 expression by phagocyte SIRPα is a critical determinant of T- and NK-cell homeostasis under steady-state conditions in vivo.


Subject(s)
Antigens, Differentiation/metabolism , CD47 Antigen/metabolism , Homeostasis , Killer Cells, Natural/metabolism , Receptors, Immunologic/metabolism , T-Lymphocytes/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Interleukin Receptor Common gamma Subunit/metabolism , Killer Cells, Natural/cytology , Kinetics , Lymphopoiesis , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Protein Binding , Receptors, Interleukin-2/deficiency , Receptors, Interleukin-2/metabolism , Spleen/cytology , Spleen/immunology , Survival Analysis , T-Lymphocytes/cytology , Thymus Gland/metabolism , Transplantation, Heterologous
14.
AIDS Res Hum Retroviruses ; 26(7): 735-46, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20624075

ABSTRACT

Humanized Rag2(-/-)gamma(c)(-/-) mice (Hu-DKO mice) become populated with functional human T cells, B cells, and dendritic cells following transplantation with human hematopoietic stem cells (HSC) and represent an improved model for studying HIV infection in vivo. In the current study we demonstrated that intrasplenic inoculation of hu-DKO mice with HIV-1 initiated a higher level of HIV infection than intravenous or intraperitoneal inoculation, associated with a reciprocal decrease in peripheral CD4(+) T cells and increase in peripheral CD8(+) T cells. HIV infection by intrasplenic injection increased serum levels of human IgG and IgM including human IgM and IgG specific for HIV-1 gp120. There was a significant inverse correlation between the level of HIV-1 infection and the extent of CD4(+) T cell depletion. Highly active antiretroviral therapy (HAART) initiated 1 week after HIV-1 inoculation markedly suppressed HIV-1 infection and prevented CD4(+) T cell depletion. Taken together, these findings demonstrate that intrasplenic injection of hu-DKO mice with HIV is a more efficient route of HIV infection than intravenous or intraperitoneal injection and generates increased infection associated with an increased anti-HIV humoral response. This animal model can serve as a valuable in vivo model to study the efficacy of anti-HIV therapies.


Subject(s)
Anti-HIV Agents/therapeutic use , Antiretroviral Therapy, Highly Active/methods , DNA-Binding Proteins/deficiency , Disease Models, Animal , HIV Infections/drug therapy , Receptors, Interleukin-2/deficiency , Animals , Animals, Newborn , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Antibodies/blood , HIV-1/immunology , Hematopoietic Stem Cell Transplantation , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Mice , Mice, Knockout
15.
J Biomed Biotechnol ; 2010: 740619, 2010.
Article in English | MEDLINE | ID: mdl-20625413

ABSTRACT

IL-2 production is decreased in systemic lupus erythematosus (SLE) patients and affects T cell function and other aspects of host immunity. Transcription factors regulating IL-2 production behave aberrantly in SLE T cells. In addition to IL-2 dysregulation, other IL-2 family members (IL-15 and IL-21) are abnormally expressed in SLE. Decreased IL-2 production in SLE patients leads to many immune defects such as decreased T(reg) production, decreased activation-induced cell death (AICD), and decreased cytotoxicity. IL-2 deficiency results in systemic dysregulation of host immune responses in patients suffering from SLE disease.


Subject(s)
Immunity/immunology , Interleukin-2/immunology , Lupus Erythematosus, Systemic/immunology , Animals , Cell Death , Disease Models, Animal , Humans , Interleukin-2/biosynthesis , Interleukin-2/deficiency , Lymphocyte Activation/immunology , Mice , Receptors, Interleukin-2/deficiency , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
16.
J Immunol ; 184(12): 7082-91, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20495069

ABSTRACT

Stable engraftment of human lymphoid tissue in NOD/scid-IL-2Rgammacnull mice after CD34+ hematopoietic stem cell reconstitution permits the evaluation of ongoing HIV-1 infection for weeks to months. We demonstrate that HIV-1-infected rodents develop virus-specific cellular immune responses. CD8+ cell depletion, 2 or 5-7 wk after viral infection, resulted in a significant increase of HIV-1 load, robust immune cell activation, and cytopathology in lymphoid tissues but preserved CD4/CD8 double-positive thymic T cell pools. Human CD8+ cells reappeared in circulation as early as 2-3 wk. These data support a role of CD8+ cells in viral surveillance and the relevance of this humanized mouse model for the studies of HIV-1 pathobiology and virus-specific immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , Animals , Cell Separation , Cord Blood Stem Cell Transplantation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Receptors, Interleukin-2/deficiency , Receptors, Interleukin-2/genetics
17.
Transplantation ; 89(5): 527-36, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20134397

ABSTRACT

BACKGROUND: Transplantation of human skin on immunodeficient mice that support engraftment with functional human immune systems would be an invaluable tool for investigating mechanisms involved in wound healing and transplantation. Nonobese diabetic (NOD)-scid interleukin-2 gamma chain receptor (NSG) readily engraft with human immune systems, but human skin graft integrity is poor. In contrast, human skin graft integrity is excellent on CB17-scid bg (SCID.bg) mice, but they engraft poorly with human immune systems. METHODS: Human skin grafts transplanted onto immunodeficient NSG, SCID.bg, and other immunodeficient strains were evaluated for graft integrity, preservation of graft endothelium, and their ability to be rejected after engraftment of allogeneic peripheral blood mononuclear cells. RESULTS: Human skin transplanted onto NSG mice develops an inflammatory infiltrate, consisting predominately of host Gr1(+) cells, that is detrimental to the survival of human endothelium in the graft. Treatment of graft recipients with anti-Gr1 antibody reduces this cellular infiltrate, preserves graft endothelium, and promotes wound healing, tissue development, and graft remodeling. Excellent graft integrity of the transplanted skin includes multilayered stratified human epidermis, well-developed human vasculature, human fibroblasts, and passenger leukocytes. Injection of unfractionated, CD4 or CD8 allogeneic human peripheral blood mononuclear cell induces a rapid destruction of the transplanted skin graft. CONCLUSIONS: NSG mice treated with anti-Gr1 antibody provide a model optimized for both human skin graft integrity and engraftment of a functional human immune system. This model provides the opportunity to investigate mechanisms orchestrating inflammation, wound healing, revascularization, tissue remodeling, and allograft rejection and can provide guidance for improving outcomes after clinical transplantation.


Subject(s)
Graft Rejection/pathology , Receptors, Interleukin-2/deficiency , Skin Transplantation/methods , Animals , Antigens, CD/analysis , Erythrocytes/physiology , Flow Cytometry , Humans , Leukocyte Common Antigens/analysis , Leukocyte Transfusion , Mice , Mice, Inbred NOD , Mice, SCID , Skin Transplantation/pathology , Spleen/pathology , Transplantation, Heterologous , Transplantation, Homologous , Wound Healing
18.
J Immunol ; 184(5): 2272-80, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20130214

ABSTRACT

Little is known about target organ-infiltrating NK cells in type 1 diabetes and other autoimmune diseases. In this study, we identified NK cells with a unique phenotype in the pancreas of NOD mice. Pancreatic NK cells, localized to the endocrine and exocrine parts, were present before T cells during disease development and did not require T cells for their infiltration. Furthermore, NK cells, or NK cell precursors, from the spleen could traffic to the pancreas, where they displayed the pancreatic phenotype. Pancreatic NK cells from other mouse strains shared phenotypic characteristics with pancreatic NK cells from NOD mice, but displayed less surface killer cell lectin-like receptor G1, a marker for mature NK cells that have undergone proliferation, and also did not proliferate to the same extent. A subset of NOD mouse pancreatic NK cells produced IFN-gamma spontaneously, suggesting ongoing effector responses. However, most NOD mouse pancreatic NK cells were hyporesponsive compared with spleen NK cells, as reflected by diminished cytokine secretion and a lower capacity to degranulate. Interestingly, such hyporesponsiveness was not seen in pancreatic NK cells from the nonautoimmune strain C57BL/6, suggesting that this feature is not a general property of pancreatic NK cells. Based on our data, we propose that NK cells are sentinel cells in a normal pancreas. We further speculate that during inflammation, pancreatic NK cells initially mediate proinflammatory effector functions, potentially contributing to organ-specific autoimmunity, but later become hyporesponsive because of exhaustion or regulation.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Killer Cells, Natural/immunology , Pancreas, Exocrine/immunology , Animals , Cell Proliferation , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Female , Flow Cytometry , Fluorescent Antibody Technique , Immunophenotyping , Islets of Langerhans/metabolism , Killer Cells, Natural/metabolism , Macrophage-1 Antigen/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Pancreas, Exocrine/metabolism , Receptors, Interleukin-2/deficiency , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/immunology , Species Specificity , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
19.
J Immunol ; 181(1): 225-34, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18566388

ABSTRACT

The IL-2/IL-2R interaction is important for development and peripheral homeostasis of T regulatory (Treg) cells. IL-2- and IL-2R-deficient mice are not completely devoid of Foxp3+ cells, but rather lack population of mature CD4+CD25+Foxp3high Treg cells and contain few immature CD4+CD25-Foxp3low T cells. Interestingly, common gamma chain (gammac) knockout mice have been shown to have a near complete absence of Foxp3+ Treg cells, including the immature CD25-Foxp3low subset. Therefore, other gammac-cytokine(s) must be critically important during thymic development of CD4+CD25+Foxp3+ Treg cells apart from the IL-2. The present study was undertaken to determine whether the gammac-cytokines IL-7 or IL-15 normally contribute to expression of Foxp3 and Treg cell production. These studies revealed that mice double deficient in IL-2Rbeta and IL-7Ralpha contained a striking lack in the CD4+Foxp3+ population and the Treg cell defect recapitulated the gammac knockout mice. In the absence of IL-7R signaling, IL-15/IL-15R interaction is dispensable for the production of CD4+CD25+Foxp3+ Treg cells, indicating that normal thymic Treg cell production likely depends on signaling through both IL-2 and IL-7 receptors. Selective thymic reconstitution of IL-2Rbeta in mice double deficient in IL-2Rbeta and IL-7Ralpha established that IL-2Rbeta is dominant and sufficient to restore production of Treg cells. Furthermore, the survival of peripheral CD4+Foxp3low cells in IL-2Rbeta-/- mice appears to depend upon IL-7R signaling. Collectively, these data indicate that IL-7R signaling contributes to Treg cell development and peripheral homeostasis.


Subject(s)
Receptors, Interleukin-7/immunology , Receptors, Interleukin-7/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Cell Differentiation/immunology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Homeostasis/immunology , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-7/metabolism , Mice , Mice, Knockout , Receptors, Interleukin-2/deficiency , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/immunology , Receptors, Interleukin-2/metabolism , Receptors, Interleukin-7/deficiency , Receptors, Interleukin-7/genetics , T-Lymphocytes, Regulatory/cytology , Thymus Gland/metabolism
20.
Immunol Rev ; 226: 19-28, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19161413

ABSTRACT

Interleukin-2 (IL-2) has multiple, sometimes opposing, functions during an inflammatory response. It is a potent inducer of T-cell proliferation and T-helper 1 (Th1) and Th2 effector T-cell differentiation and provides T cells with a long-lasting competitive advantage resulting in the optimal survival and function of memory cells. In a regulatory role, IL-2 is important for the development, survival, and function of regulatory T cells, it enhances Fas-mediated activation-induced cell death, and it inhibits the development of inflammatory Th17 cells. Thus, in its dual and contrasting functions, IL-2 contributes to both the induction and the termination of inflammatory immune responses.


Subject(s)
Inflammation/immunology , Interleukin-2/immunology , Receptors, Interleukin-2/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytokines/immunology , Cytokines/metabolism , Humans , Inflammation/metabolism , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Interleukin-2/deficiency , Receptors, Interleukin-2/deficiency , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...