Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Life Sci ; 254: 117813, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32428597

ABSTRACT

AIMS: This study aimed to investigate the effect of lymphocytes in wound healing and the underlying mechanisms, in diabetic and non-diabetic mice, using Balb/c recombination activating gene (Rag)-2 and interleukin 2 receptor gamma (IL-2Rγ) double knockout (KO) (RAG2-/- IL-2Rγ-/-) mice. MAIN METHODS: Wound healing in vivo was performed in control and STZ-induced diabetic mice, in both KO and WT mice. Inflammation and ROS production were evaluated by immunofluorescence microscopy analysis, antioxidant enzymes and angiogenesis were evaluated by quantitative PCR and immunofluorescence microscopy analysis, and wound closure kinetics evolution was evaluated by measurement of acetate tracing of the wound area. KEY FINDINGS: Wound closure was significantly delayed in KO mice, where the M1/M2 macrophage ratio and basal ROS levels were significantly increased, while antioxidant defenses and angiogenesis were significantly decreased. Moreover, the expected increase in matrix metallopeptidase (MMP)-9 protein levels in diabetic conditions was not observed in KO mice, suggesting that the mechanisms leading to the increase in MMP-9 observed in diabetic wounds may in part be lymphocyte-dependent. SIGNIFICANCE: Our results indicate that lack of lymphocytes compromises wound healing independent of diabetes. The lack of these cells, even in non-diabetic mice, mimics the phenotype observed in wounds under diabetic conditions. Moreover, the combination of diabetes and the lack of lymphocytes, further impair the wound healing conditions, indicating that when the innate regulatory function is lost in these KO mice, excessive M1 polarization, poor angiogenesis and impaired wound healing are worsen.


Subject(s)
DNA-Binding Proteins/physiology , Diabetes Mellitus, Experimental/physiopathology , Interleukin Receptor Common gamma Subunit/physiology , Lymphocytes/physiology , Neovascularization, Physiologic/physiology , Wound Healing/physiology , Animals , DNA-Binding Proteins/genetics , Inflammation/metabolism , Interleukin Receptor Common gamma Subunit/genetics , Macrophage Activation/physiology , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Reactive Oxygen Species/metabolism , Skin/blood supply , Skin/metabolism
2.
J Leukoc Biol ; 107(1): 27-42, 2020 01.
Article in English | MEDLINE | ID: mdl-31378988

ABSTRACT

This work sought to confirm the human-like expression of exhaustion and senescence markers in a mouse model with a humanized immune system (HIS): the Balb/c Rag2KO IL2rgcKO SirpαNOD Flk2KO HLA-A2HHD (BRGSF-A2) mouse reconstituted with human CD34+ cord blood cells. With regard to senescence markers, the percentage of CD57+ T cells was higher in the bone marrow (BM) than in the spleen or blood. The same was true for KLRG1+ hCD8+ T cells. With regard to exhaustion markers, the percentage of programmed death 1 (PD-1+ ) T cells was higher in the BM than in the spleen or blood; the same was true for TIGIT+ hCD4+ cells. These tissue-specific differences were related to both higher proportions of memory T cells in BM and intrinsic differences in expression within the memory fraction. In blood samples from HIS mice and healthy human donors (HDs), we found that the percentage of KLRG1+ cells among hCD8+ T cells was lower in HIS compared to HDs. The opposite was true for CD4+ T cells. Unexpectedly, a high frequency of KLRG1+ cells was observed among naive T cells in HIS mice. CD57 expression on T cells was similar in blood samples from HIS mice and HDs. Likewise, PD-1 expression was similar in the two systems, although a relatively low proportion of HIS hCD4+ T cells expressed TIGIT. The BRGSF-A2 HIS mouse's exhaustion and senescence profile was tissue specific and relatively human like; hence, this mouse might be a valuable tool for determining the preclinical efficacy of immunotherapies.


Subject(s)
Biomarkers/analysis , Cellular Senescence , DNA-Binding Proteins/physiology , HLA-A2 Antigen/physiology , Interleukin Receptor Common gamma Subunit/physiology , Receptors, Immunologic/physiology , T-Lymphocytes/immunology , fms-Like Tyrosine Kinase 3/physiology , Adult , Aged , Animals , Female , Healthy Volunteers , Humans , Leukocytes , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Knockout , Middle Aged , Receptors, Immunologic/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
3.
Adv Exp Med Biol ; 1172: 1-19, 2019.
Article in English | MEDLINE | ID: mdl-31628649

ABSTRACT

The common γ chain (γc) family of hematopoietic cytokines consists of six distinct four α-helix bundle soluble ligands that signal through receptors which include the shared γc subunit to coordinate a wide range of physiological processes, in particular, those related to innate and adaptive immune function. Since the first crystallographic structure of a γc family cytokine/receptor signaling complex (the active Interleukin-2 [IL-2] quaternary complex) was determined in 2005 [1], tremendous progress has been made in the structural characterization of this protein family, transforming our understanding of the molecular mechanisms underlying immune activity. Although many conserved features of γc family cytokine complex architecture have emerged, distinguishing details have been observed for individual cytokine complexes that rationalize their unique functional properties. Much work remains to be done in the molecular characterization of γc family signaling, particularly with regard to intracellular activation events, and looking forward, new technologies in structural biophysics will offer further insight into the biology of cytokine signaling to inform the design of targeted therapeutics for treatment of immune-linked diseases such as cancer, infection, and autoimmune disorders.


Subject(s)
Interleukin Receptor Common gamma Subunit , Signal Transduction , Animals , Humans , Interleukin Receptor Common gamma Subunit/chemistry , Interleukin Receptor Common gamma Subunit/physiology , Interleukin-2/chemistry , Interleukin-2/physiology , Receptors, Cytokine/metabolism , Signal Transduction/physiology
4.
Int J Cancer ; 143(5): 1212-1223, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29577276

ABSTRACT

Previous studies have shown that soluble common γ-chain (sγc) modulates CD4+ T cell immunity with antagonistic functions in γc cytokine signaling. However, the role of sγc in functional properties of effector CD8+ T cells has not been fully defined. In this study, we report a new mechanism by which the anti-tumor activity of mouse CD8+ T cells is suppressed in sγc of their own producing. While sγc significantly inhibits cytotoxicity of CD8+ T cells, blocking sγc production by genetic modification leads to potentiated effector function of CD8+ T cells, establishing persistent CD8+ T cells. This is due to the modulation of IL-2 and IL-15 signaling, which is required for expansion and survival of CD8+ T cells as well as for optimal cytotoxic activity. More efficient management of tumor growth was achieved by an adoptive transfer of sγc-deficient CD8+ T cells than that of wild-type or sγc-overexpressing CD8+ T cells. Blocking of IL-2 and IL-15 signaling by sγc attenuates the capacity of CD8+ T cells to mount an optimal response to the tumor, with both quantitative and qualitative effects on antigen-specific CD8+ T cells. These results could have a critical implication for the generation and survival of optimal effector T cells for adoptive immunotherapy of cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy , Interleukin Receptor Common gamma Subunit/physiology , Interleukin-15/immunology , Interleukin-2/immunology , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Signal Transduction , Tumor Cells, Cultured
5.
Reprod Biol Endocrinol ; 14(1): 74, 2016 Nov 03.
Article in English | MEDLINE | ID: mdl-27809915

ABSTRACT

BACKGROUND: Pigs with SCID can be a useful model in regenerative medicine, xenotransplantation, and cancer cell transplantation studies. Utilizing genome editing technologies such as CRISPR/Cas9 system allows us to generate genetically engineered pigs at a higher efficiency. In this study, we report generation and phenotypic characterization of IL2RG knockout female pigs produced through combination of CRISPR/Cas9 system and SCNT. As expected, pigs lacking IL2RG presented SCID phenotype. METHODS: First, specific CRISPR/Cas9 systems targeting IL2RG were introduced into developing pig embryos then the embryos were transferred into surrogates. A total of six fetuses were obtained from the embryo transfer and fetal fibroblast cell lines were established. Then IL2RG knockout female cells carrying biallelic genetic modification were used as donor cells for SCNT, followed by embryo transfer. RESULTS: Three live cloned female piglets carrying biallelic mutations in IL2RG were produced. All cloned piglets completely lacked thymus and they had a significantly reduced level of mature T, B and NK cells in their blood and spleen. CONCLUSIONS: Here, we generated IL2RG knockout female pigs showing phenotypic characterization of SCID. This IL2RG knockout female pigs will be a promising model for biomedical and translational research.


Subject(s)
Interleukin Receptor Common gamma Subunit/genetics , Models, Animal , Severe Combined Immunodeficiency/veterinary , Swine Diseases/genetics , Alleles , Animals , Female , Gene Knockout Techniques , Genetic Engineering , Interleukin Receptor Common gamma Subunit/physiology , Severe Combined Immunodeficiency/genetics , Swine
6.
Am J Transplant ; 16(2): 440-53, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26550777

ABSTRACT

CD8(+)/TCR(-) facilitating cells (FCs) in mouse bone marrow (BM) significantly enhance engraftment of hematopoietic stem/progenitor cells (HSPCs). Human FC phenotype and mechanism of action remain to be defined. We report, for the first time, the phenotypic characterization of human FCs and correlation of phenotype with function. Approximately half of human FCs are CD8(+)/TCR(-)/CD56 negative (CD56(neg)); the remainder are CD8(+)/TCR(-)/CD56 bright (CD56(bright)). The CD56(neg) FC subpopulation significantly promotes homing of HSPCs to BM in nonobese diabetic/severe combined immunodeficiency/IL-2 receptor γ-chain knockout mouse recipients and enhances hematopoietic colony formation in vitro. The CD56(neg) FC subpopulation promotes rapid reconstitution of donor HSPCs without graft-versus-host disease (GVHD); recipients of CD56(bright) FCs plus HSPCs exhibit low donor chimerism early after transplantation, but the level of chimerism significantly increases with time. Recipients of HSPCs plus CD56(neg) or CD56(bright) FCs showed durable donor chimerism at significantly higher levels in BM. The majority of both FC subpopulations express CXCR4. Coculture of CD56(bright) FCs with HSPCs upregulates cathelicidin and ß-defensin 2, factors that prime responsiveness of HSPCs to stromal cell-derived factor 1. Both FC subpopulations significantly upregulated mRNA expression of the HSPC growth factors and Flt3 ligand. These results indicate that human FCs exert a direct effect on HSPCs to enhance engraftment. Human FCs offer a potential regulatory cell-based therapy for enhancement of engraftment and prevention of GVHD.


Subject(s)
CD8 Antigens/metabolism , Graft vs Host Disease/immunology , Hematopoietic Stem Cells/immunology , Interleukin Receptor Common gamma Subunit/physiology , Receptors, Antigen, T-Cell/metabolism , Animals , Apoptosis , Blotting, Western , Cells, Cultured , Graft vs Host Disease/metabolism , Hematopoietic Stem Cells/metabolism , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Models, Animal , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tissue Donors , Transplantation Chimera
7.
Oncotarget ; 6(31): 32039-60, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26451606

ABSTRACT

Cancer stem cells (CSCs) play major roles in cancer initiation, progression, and metastasis. It is evident from growing reports that PI3K/Akt/mTOR and Sonic Hedgehog (Shh) signaling pathways are aberrantly reactivated in pancreatic CSCs. Here, we examined the efficacy of combining NVP-LDE-225 (PI3K/mTOR inhibitor) and NVP-BEZ-235 (Smoothened inhibitor) on pancreatic CSCs characteristics, microRNA regulatory network, and tumor growth. NVP-LDE-225 co-operated with NVP-BEZ-235 in inhibiting pancreatic CSC's characteristics and tumor growth in mice by acting at the level of Gli. Combination of NVP-LDE-225 and NVP-BEZ-235 inhibited self-renewal capacity of CSCs by suppressing the expression of pluripotency maintaining factors Nanog, Oct-4, Sox-2 and c-Myc, and transcription of Gli. NVP-LDE-225 co-operated with NVP-BEZ-235 to inhibit Lin28/Let7a/Kras axis in pancreatic CSCs. Furthermore, a superior interaction of these drugs was observed on spheroid formation by pancreatic CSCs isolated from Pankras/p53 mice. The combination of these drugs also showed superior effects on the expression of proteins involved in cell proliferation, survival and apoptosis. In addition, NVP-LDE-225 co-operated with NVP-BEZ-235 in inhibiting EMT through modulation of cadherin, vimentin and transcription factors Snail, Slug and Zeb1. In conclusion, these data suggest that the combined inhibition of PI3K/Akt/mTOR and Shh pathways may be beneficial for the treatment of pancreatic cancer.


Subject(s)
Enzyme Inhibitors/pharmacology , Hedgehog Proteins/metabolism , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Blotting, Western , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Profiling , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/genetics , Humans , Immunoenzyme Techniques , Interleukin Receptor Common gamma Subunit/physiology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Vet Immunol Immunopathol ; 157(3-4): 131-41, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24368085

ABSTRACT

To study the canine immune system we generated a mouse model engrafted with canine lymphocytes using NOD SCID IL2R common gamma chain -/- (NSG) mice as recipients (Ca-PBL-SCID). Engraftment of canine peripheral blood lymphocytes (PBLs) was determined post-injection with 10(7) peripheral blood mononuclear cells (PBMCs) into irradiated NSG mice using flow cytometry and fluorescently labeled antibodies specific to canine helper T cells (CD45(+) CD4(+)), cytotoxic lymphocytes (CD45(+) CD8(+)), regulatory T cells (CD45(+) CD4(+) Foxp3(+)), and B cells (CD45(+) Ig(+) CD21lo). Canine CD45(+) lymphocytes were detectable as early as day 1 in the peritoneal cavity, and beginning at 9 days in the blood, bone marrow, and spleen. CD4(+) T cells, of which Foxp-3(+) CD25hi cells constituted a minor percentage, were the predominant lymphocyte population at 9 days post engraftment contrasting with increasing proportions of CD8(+) CTL's and Ig(+) B cells beginning at 16 days. Canine immunoglobulin was initially detected in the serum of Ca-PBL-SCID mice at 9 days post-engraftment and peaked in concentration at day 36. From day 28 to 52 post-engraftment 30% of the Ca-PBL-SCID mice became markedly anemic and thrombocytopenic, yet gross and histopathologic examination of bone marrow, kidneys, spleen, liver, and intestine revealed no obvious lesions. Blood smear evaluation revealed agglutination of mature red blood cells, reticulocytes and a regenerative anemia. These findings demonstrate that NSG mice are capable of engraftment of canine PBLs yet develop graft versus host disease similar to Hu-PBL-SCID mice.


Subject(s)
Dogs/immunology , Interleukin Receptor Common gamma Subunit/physiology , Lymphocytes/immunology , Anemia, Hemolytic/etiology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Graft vs Host Disease/immunology , Heterografts , Mice , Mice, Inbred NOD , Mice, SCID , Radiation Tolerance
9.
Exp Hematol ; 42(1): 28-38.e1-2, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24120693

ABSTRACT

Current strategies in cellular immunotherapy of cancer and viral infections include the adoptive transfer of T cell receptor (TCR) and chimeric antigen receptor engineered T cells. When using transient RNA expression systems in clinical studies, multiple infusions with receptor-redirected T cells appear necessary. However, in allogeneic hematopoietic stem-cell transplantation, repeated transfer of donor-derived T cells increases the risk of alloreactive graft-versus-host disease. We investigated naive-derived (TN), memory-derived (TM), and Epstein Barr virus-specific (TEBV) CD8(+) T cell subsets for alloreactivity upon redirection with RNA encoding a cytomegalovirus-specific model TCR. We observed that alloreactivity to human leukocyte antigen (HLA)-mismatched hematopoietic cells developed at much stronger levels in TN compared with TM or TEBV populations in cytokine-release and cytotoxicity assays. Cytomegalovirus-specific effector function was higher in TCR-transfected TEBV and TM over TN cells. To measure alloreactivity in vivo, we reconstituted NOD/SCID/IL-2Rγc(null) mice with human CD34(+) stem cells and adoptively transferred them with CD8(+) T cell subsets previously stimulated against cells of the HLA-mismatched stem-cell donor. TN cells showed a significant ability to eliminate CD34-derived hematopoietic cells, which was not found with TM and TEBV cells. This reduced alloreactive potential along with strong effector function upon receptor RNA engineering makes CD8(+) memory and EBV-specific T cells advantageous tools in adoptive immunotherapy after allogeneic transplantation.


Subject(s)
Antigens, CD34/analysis , CD8-Positive T-Lymphocytes/immunology , Hematopoietic Stem Cell Transplantation , Herpesvirus 4, Human/immunology , Immunologic Memory , Interleukin Receptor Common gamma Subunit/physiology , T-Lymphocytes/immunology , Animals , Histocompatibility Testing , Humans , Immunotherapy, Adoptive , Mice , Mice, SCID , Transplantation, Homologous
10.
PLoS One ; 8(10): e77780, 2013.
Article in English | MEDLINE | ID: mdl-24204962

ABSTRACT

An estimated 34 million people are living with HIV worldwide (UNAIDS, 2012), with the number of infected persons rising every year. Increases in HIV prevalence have resulted not only from new infections, but also from increases in the survival of HIV-infected persons produced by effective anti-retroviral therapies. Augmentation of anti-viral immune responses may be able to further increase the survival of HIV-infected persons. One strategy to augment these responses is to reinvigorate exhausted anti-HIV immune cells present in chronically infected persons. The PD-1-PD-L1 pathway has been implicated in the exhaustion of virus-specific T cells during chronic HIV infection. Inhibition of PD-1 signaling using blocking anti-PD-1 antibodies has been shown to reduce simian immunodeficiency virus (SIV) loads in monkeys. We now show that PD-1 blockade can improve control of HIV replication in vivo in an animal model. BLT (Bone marrow-Liver-Thymus) humanized mice chronically infected with HIV-1 were treated with an anti-PD-1 antibody over a 10-day period. The PD-1 blockade resulted in a very significant 45-fold reduction in HIV viral loads in humanized mice with high CD8(+) T cell expression of PD-1, compared to controls at 4 weeks post-treatment. The anti-PD-1 antibody treatment also resulted in a significant increase in CD8(+) T cells. PD-1 blockade did not affect T cell expression of other inhibitory receptors co-expressed with PD-1, including CD244, CD160 and LAG-3, and did not appear to affect virus-specific humoral immune responses. These data demonstrate that inhibiting PD-1 signaling can reduce HIV viral loads in vivo in the humanized BLT mouse model, suggesting that blockade of the PD-1-PD-L1 pathway may have therapeutic potential in the treatment of patients already infected with the AIDS virus.


Subject(s)
Antibodies, Blocking/pharmacology , CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Viral Load/drug effects , Animals , Blotting, Western , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , HIV Infections/virology , HIV-1/drug effects , Humans , Interleukin Receptor Common gamma Subunit/physiology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Programmed Cell Death 1 Receptor/immunology
11.
Cancer Res ; 73(18): 5810-20, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23928995

ABSTRACT

Basal-type triple-negative breast cancers (TNBC) are aggressive and difficult to treat relative to luminal-type breast cancers. TNBC often express abundant Met receptors and are enriched for transcriptional targets regulated by hypoxia-inducible factor-1α (HIF-1α), which independently predict cancer relapse and increased risk of metastasis. Brk/PTK6 is a critical downstream effector of Met signaling and is required for hepatocyte growth factor (HGF)-induced cell migration. Herein, we examined the regulation of Brk by HIFs in TNBC in vitro and in vivo. Brk mRNA and protein levels are upregulated strongly in vitro by hypoxia, low glucose, and reactive oxygen species. In HIF-silenced cells, Brk expression relied upon both HIF-1α and HIF-2α, which we found to regulate BRK transcription directly. HIF-1α/2α silencing in MDA-MB-231 cells diminished xenograft growth and Brk reexpression reversed this effect. These findings were pursued in vivo by crossing WAP-Brk (FVB) transgenic mice into the MET(Mut) knockin (FVB) model. In this setting, Brk expression augmented MET(Mut)-induced mammary tumor formation and metastasis. Unexpectedly, tumors arising in either MET(Mut) or WAP-Brk × MET(Mut) mice expressed abundant levels of Sik, the mouse homolog of Brk, which conferred increased tumor formation and decreased survival. Taken together, our results identify HIF-1α/2α as novel regulators of Brk expression and suggest that Brk is a key mediator of hypoxia-induced breast cancer progression. Targeting Brk expression or activity may provide an effective means to block the progression of aggressive breast cancers.


Subject(s)
Breast Neoplasms/pathology , Breast/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/pathology , Neoplasm Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Breast/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/mortality , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/mortality , Cell Proliferation , Chromatin Immunoprecipitation , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoenzyme Techniques , Interleukin Receptor Common gamma Subunit/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Neoplasm Proteins/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
13.
Leukemia ; 27(11): 2139-48, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23568148

ABSTRACT

Chemotherapy remains mainly used for the treatment of acute myeloid leukemia (AML). However, in the past 3 decades limited progress has been achieved in improving the long-term disease-free survival. Therefore the development of more effective drugs for AML represents a high level of priority. F14512 combines an epipodophyllotoxin core targeting topoisomerase II with a spermine moiety introduced as a cell delivery vector. The polyamine moiety facilitates F14512 selective uptake by tumour cells via the polyamine transport system, a machinery overactivated in cancer cells. F14512 has been characterized as a potent drug candidate and is currently in Phase I clinical trials. Here, we demonstrated marked survival benefit and therapeutic efficacy of F14512 treatments in a series of human AML models, established either from AML cell lines or from patient AML samples. Furthermore, we reported in vitro synergistic anti-leukemic effects of F14512 in combination with cytosine arabinoside (Ara-C), doxorubicin, gemcitabine, bortezomib or SAHA. In vivo combination of suboptimal doses of F14512 with Ara-C also resulted in enhanced anti-leukemic activity. We further showed that F14512 triggered both senescence and apoptosis in vivo in primary AML models, but not autophagy. Overall, these results support the clinical development in onco-hematology of this novel promising drug candidate.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Fetal Blood/drug effects , Interleukin Receptor Common gamma Subunit/physiology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Animals , Blotting, Western , Boronic Acids/administration & dosage , Bortezomib , Cytarabine/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Doxorubicin/administration & dosage , Fetal Blood/cytology , Flow Cytometry , Humans , Hydroxamic Acids/administration & dosage , Immunoenzyme Techniques , Mice , Mice, Inbred NOD , Mice, SCID , Podophyllotoxin/administration & dosage , Podophyllotoxin/analogs & derivatives , Pyrazines/administration & dosage , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured , Vorinostat , Xenograft Model Antitumor Assays , Gemcitabine
14.
J Hematol Oncol ; 6: 21, 2013 Mar 16.
Article in English | MEDLINE | ID: mdl-23497456

ABSTRACT

BACKGROUND: Activating mutations [internal tandem duplication (ITD)] or overexpression of the FMS-like tyrosine kinase receptor-3 (FLT3) gene are associated with poor outcome in acute myeloid leukemia (AML) patients, underscoring the need for novel therapeutic approaches. The natural product silvestrol has potent antitumor activity in several malignancies, but its therapeutic impact on distinct molecular high-risk AML subsets remains to be fully investigated. We examined here the preclinical activity of silvestrol in FLT3-ITD and FLT3 wild-type (wt) AML. METHODS: Silvestrol in vitro anti-leukemic activity was examined by colorimetric cell viability assay, colony-forming and flow cytometry assays assessing growth inhibition and apoptosis, respectively. Pharmacological activity of silvestrol on FLT3 mRNA translation, mRNA and protein expression was determined by RNA-immunoprecipitation, qRT-PCR and immunoblot analyses, respectively. Silvestrol in vivo efficacy was investigated using MV4-11 leukemia-engrafted mice. RESULTS: Silvestrol shows antileukemia activity at nanomolar concentrations both in FLT3-wt overexpressing (THP-1) and FLT3-ITD (MV4-11) expressing AML cell lines (IC50 = 3.8 and 2.7 nM, respectively) and patients' primary blasts [IC50 = ~12 nM (FLT3-wt) and ~5 nM (FLT3-ITD)]. Silvestrol increased apoptosis (~4fold, P = 0.0001), and inhibited colony-formation (100%, P < 0.0001) in primary blasts. Silvestrol efficiently inhibited FLT3 translation reducing FLT3 protein expression by 80-90% and decreased miR-155 levels (~60%), a frequently co-regulated onco-miR in FLT3-ITD-positive AML. The median survival of silvestrol-treated vs vehicle-treated mice was 63 vs 29 days post-engraftment, respectively (P < 0.0001). CONCLUSIONS: Silvestrol exhibits significant in vivo and in vitro antileukemic activities in AML through a novel mechanism resulting in inhibition of FLT3 and miR-155 expression. These encouraging results warrant a rapid translation of silvestrol for clinical testing in AML.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Interleukin Receptor Common gamma Subunit/physiology , Leukemia, Myeloid, Acute/drug therapy , MicroRNAs/antagonists & inhibitors , Triterpenes/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Apoptosis/drug effects , Blast Crisis/drug therapy , Blast Crisis/metabolism , Blast Crisis/pathology , Blotting, Western , Cell Proliferation/drug effects , Flow Cytometry , Humans , In Vitro Techniques , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
15.
Haematologica ; 98(7): 1030-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23403317

ABSTRACT

Mutations in the common gamma chain (γc, CD132, encoded by the IL2RG gene) can lead to B(+)T(-)NK(-) X-linked severe combined immunodeficiency, as a consequence of unresponsiveness to γc-cytokines such as interleukins-2, -7 and -15. Hypomorphic mutations in CD132 may cause combined immunodeficiencies with a variety of clinical presentations. We analyzed peripheral blood mononuclear cells of a 6-year-old boy with normal lymphocyte counts, who suffered from recurrent pneumonia and disseminated mollusca contagiosa. Since proliferative responses of T cells and NK cells to γc -cytokines were severely impaired, we performed IL2RG gene analysis, showing a heterozygous mutation in the presence of a single X-chromosome. Interestingly, an IL2RG reversion to normal predominated in both naïve and antigen-primed CD8(+) T cells and increased over time. Only the revertant CD8(+) T cells showed normal expression of CD132 and the various CD8(+) T cell populations had a different T-cell receptor repertoire. Finally, a fraction of γδ(+) T cells and differentiated CD4(+)CD27(-) effector-memory T cells carried the reversion, whereas NK or B cells were repeatedly negative. In conclusion, in a patient with a novel IL2RG mutation, gene-reverted CD8(+) T cells accumulated over time. Our data indicate that selective outgrowth of particular T-cell subsets may occur following reversion at the level of committed T progenitor cells.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Interleukin Receptor Common gamma Subunit/genetics , Mutation/genetics , Severe Combined Immunodeficiency/genetics , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation , Child , Humans , Interleukin Receptor Common gamma Subunit/physiology , Lymphocyte Count , Male , Pedigree , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/pathology
16.
J Hematol Oncol ; 6: 10, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23343252

ABSTRACT

BACKGROUND: We investigated the utility of bioluminescence imaging (BLI) using firefly luciferase in monoclonal and polyclonal populations of leukemia cells in vitro and in vivo. METHODS: Monoclonal and polyclonal human lymphoid and myeloid leukemia cell lines transduced with firefly luciferase were used for BLI. RESULTS: Kinetics and dynamics of bioluminescence signal were cell line dependent. Luciferase expression decreased significantly over time in polyclonal leukemia cells in vitro. Transplantation of polyclonal luciferase-tagged cells in mice resulted in inconsistent signal intensity. After selection of monoclonal cell populations, luciferase activity was stable, equal kinetic and dynamic of bioluminescence intensity and strong correlation between cell number and light emission in vitro were observed. We obtained an equal development of leukemia burden detected by luciferase activity in NOD-scid-gamma mice after transplantation of monoclonal populations. CONCLUSION: The use of monoclonal leukemia cells selected for stable and equal luciferase activity is recommended for experiments in vitro and xenograft mouse models. The findings are highly significant for bioluminescence imaging focused on pre-clinical drug development.


Subject(s)
Disease Models, Animal , Leukemia, Experimental/pathology , Luciferases, Firefly/metabolism , Luminescent Measurements , Photons , Animals , Humans , Image Processing, Computer-Assisted , In Vitro Techniques , Interleukin Receptor Common gamma Subunit/physiology , Kinetics , Leukemia, Experimental/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Cells, Cultured
17.
Leukemia ; 27(1): 56-65, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22828445

ABSTRACT

The transcriptional regulator ecotropic viral integration site-1 (EVI-1) has mainly been studied for its role in myeloid malignancies, in which high EVI-1 levels are associated with particularly aggressive disease. The role of EVI-1 in lymphoid cells, however, is largely unknown. Here we show that EVI-1 is indeed expressed in lymphoid malignancies such as acute lymphoblastic leukemia (ALL) and a subset of chronic lymphocytic leukemia. Expression data from pediatric ALL further suggest that high EVI-1 levels are associated with poor prognosis. Suppression of EVI-1 expression by RNA interference reduces cell growth and enhances apoptosis sensitivity in response to various stimuli in lymphoblastic leukemia cells. At the molecular level, EVI-1 modulates expression of several apoptosis-related genes (such as BCL2, BCL-x, XIAP, NOXA, PUMA, TRAIL-R1). Furthermore, EVI-1 knockdown strongly impairs in vivo engraftment of lymphoblastic leukemia cells upon transplantation in immune-permissive NOD/SCID/IL2Rγ(null) mice, conferring a survival benefit when compared with mice transplanted with control cells. Thus, our data show that EVI-1 is expressed not only in myeloid but also in lymphoid leukemias, and contributes to the leukemogenic potential and apoptosis resistance of ALL cells.


Subject(s)
Apoptosis , DNA-Binding Proteins/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Leukemia, Myeloid, Acute/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Transcription Factors/metabolism , Adult , Animals , Blotting, Western , Case-Control Studies , Cell Cycle , Cell Proliferation , Child , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Fluorescent Antibody Technique , Humans , Interleukin Receptor Common gamma Subunit/physiology , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , MDS1 and EVI1 Complex Locus Protein , Mice , Mice, Inbred NOD , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogenes/genetics , RNA, Small Interfering/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Tumor Cells, Cultured
18.
Leukemia ; 27(1): 66-74, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22847027

ABSTRACT

Drugs that target the chief mediator of nuclear export, chromosome region maintenance 1 protein (CRM1) have potential as therapeutics for leukemia, but existing CRM1 inhibitors show variable potencies and a broad range of cytotoxic effects. Here, we report the structural analysis and antileukemic activity of a new generation of small-molecule inhibitors of CRM1. Designated selective inhibitors of nuclear export (SINE), these compounds were developed using molecular modeling to screen a small virtual library of compounds against the nuclear export signal (NES) groove of CRM1. The 2.2-Å crystal structure of the CRM1-Ran-RanBP1 complex bound to KPT-251, a representative molecule of this class of inhibitors, shows that the drug occupies part of the groove in CRM1 that is usually occupied by the NES, but penetrates much deeper into the groove and blocks CRM1-directed protein export. SINE inhibitors exhibit potent antileukemic activity, inducing apoptosis at nanomolar concentrations in a panel of 14 human acute myeloid leukemia (AML) cell lines representing different molecular subtypes of the disease. When administered orally to immunodeficient mice engrafted with human AML cells, KPT-251 had potent antileukemic activity with negligible toxicity to normal hematopoietic cells. Thus, KPT-SINE CRM1 antagonists represent a novel class of drugs that warrant further testing in AML patients.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Antineoplastic Agents/pharmacology , Karyopherins/chemistry , Leukemia, Myeloid, Acute/drug therapy , Nuclear Export Signals , Nuclear Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , ran GTP-Binding Protein/metabolism , Animals , Antineoplastic Agents/chemistry , Apoptosis , Blotting, Western , Cell Cycle , Cell Nucleus/metabolism , Cell Proliferation , Cells, Cultured , Crystallization , Crystallography, X-Ray , Female , Hematopoietic Stem Cells , Humans , Interleukin Receptor Common gamma Subunit/physiology , Karyopherins/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Nuclear Proteins/chemistry , Protein Binding , Receptors, Cytoplasmic and Nuclear/metabolism , Small Molecule Libraries , Xenograft Model Antitumor Assays , ran GTP-Binding Protein/chemistry , Exportin 1 Protein
19.
Blood ; 121(1): 107-17, 2013 Jan 03.
Article in English | MEDLINE | ID: mdl-23043071

ABSTRACT

The primary consequence of positive selection is to render thymocytes responsive to cytokines and chemokines expressed in the thymic medulla. In the present study, our main objective was to discover which cytokines could support the differentiation of positively selected thymocytes. To this end, we have developed an in vitro model suitable for high-throughput analyses of positive selection and CD8 T-cell differentiation. The model involves coculture of TCR(hi)CD5(int)CD69(-) double-positive (DP) thymocytes with peptide-pulsed OP9 cells and γc-cytokines. We report that IL-4, IL-7, and IL-21 have nonredundant effects on positively selected DP thymocytes. IL-7 signaling phosphorylates STAT5 and ERK; induces Foxo1, Klf2, and S1pr1; and supports the differentiation of classic CD8 T cells. IL-4 activates STAT6 and ERK and supports the differentiation of CD8(int)PD-L1(hi)CD44(hi)EOMES(+) innate CD8 T cells. IL-21 is produced by thymic epithelial cells and the IL-21 receptor-α is strongly induced on DP thymocytes undergoing positive selection. IL-21 signaling phosphorylates STAT3 and STAT5, but not ERK, and does not support CD8 T-cell differentiation. However, IL-21 has a unique ability to up-regulate BCL-6, expand DP thymocytes undergoing positive selection, and increase the production of mature T cells. Our data suggest that injection of recombinant IL-21 might enhance thymic output in subjects with age- or disease-related thymic atrophy.


Subject(s)
Clonal Selection, Antigen-Mediated/drug effects , Cytokines/physiology , Interleukin Receptor Common gamma Subunit/drug effects , Lymphopoiesis/drug effects , Signal Transduction/drug effects , T-Lymphocytes/cytology , Thymocytes/cytology , Thymus Gland/cytology , Animals , Atrophy , Cells, Cultured/cytology , Cells, Cultured/drug effects , Coculture Techniques , Cytokines/pharmacology , Epithelial Cells/metabolism , High-Throughput Screening Assays , Immunocompetence/drug effects , Interleukin Receptor Common gamma Subunit/physiology , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Processing, Post-Translational , Recombinant Proteins/pharmacology , STAT Transcription Factors/metabolism , Specific Pathogen-Free Organisms , Thymus Gland/drug effects , Thymus Gland/immunology , Thymus Gland/pathology
20.
Hepatobiliary Pancreat Dis Int ; 11(5): 494-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23060394

ABSTRACT

BACKGROUND: A major barrier to the clinical application of xenotransplantation as a treatment option for patients is T cell-mediated rejection. Studies based on experimental rodent models of xenograft tolerance or rejection in vivo have provided useful information about the role of T cell immune response in xenotransplantation. However not all observations seen in rodents faithfully recapitulate the human situation. This study aimed to establish a humanized mouse model of xenotransplantation, which mimics xenograft rejection in the context of the human immune system. METHODS: NOD-SCID IL2rgamma-/- mice were transplanted with neonatal porcine islet cell clusters (NICC) followed by reconstitution of human peripheral blood mononuclear cells (PBMC). Human leukocyte engraftment and islet xenograft rejection were confirmed by flow cytometric and histological analyses. RESULTS: In the absence of human PBMC, porcine NICC transplanted into NOD-SCID IL2rgamma-/- mice revealed excellent graft integrity and endocrine function. Human PBMC demonstrated a high level of engraftment in NOD-SCID IL2rgamma-/- mice. Reconstitution of NICC recipient NOD-SCID IL2rgamma-/- mice with human PBMC led to the rapid destruction of NICC xenografts in a PBMC number-dependent manner. CONCLUSIONS: Human PBMC-reconstituted NOD-SCID IL2rgamma-/- mice provide an ideal model to study human immune responses in xenotransplantation. Studies based on this humanized mouse model will provide insight for improving the outcomes of clinical xenotransplantation.


Subject(s)
Transplantation, Heterologous/immunology , Animals , Humans , Interleukin Receptor Common gamma Subunit/physiology , Islets of Langerhans Transplantation/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Models, Animal , Swine
SELECTION OF CITATIONS
SEARCH DETAIL