Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Proc Natl Acad Sci U S A ; 120(15): e2217562120, 2023 04 11.
Article in English | MEDLINE | ID: mdl-37014858

ABSTRACT

Naïve T cells and regulatory T cells, when purified, do not proliferate to the γc-cytokines IL-2, IL-7, or IL-15, despite their expression of cognate cytokine receptors. Dendritic cells (DCs) enabled the T cell proliferation to these cytokines, through cell-to-cell contact, but independent of T cell receptor stimulation. This effect lasted after separation of T cells from DCs, enabling enhanced proliferation of the T cells in DC-depleted hosts. We propose calling this a "preconditioning effect". Interestingly, IL-2 alone was sufficient to induce phosphorylation and nuclear translocation of STAT5 in T cells, but could not activate MAPK and AKT pathways and failed to induce transcription of IL-2 target genes. "Preconditioning" was necessary to activate these two pathways and induced weak Ca2+ mobilization independent of calcium release-activated channels. When preconditioning was combined with IL-2, full activation of downstream mTOR, 4E-BP1 hyperphosphorylation, and prolonged S6 phosphorylation occurred. Collectively, accessory cells provide T cell preconditioning, a unique activation mechanism, controlling cytokine-mediated proliferation of T cells.


Subject(s)
Cytokines , Interleukin Receptor Common gamma Subunit , Cytokines/metabolism , T-Lymphocytes, Regulatory/metabolism , Interleukin-2/pharmacology , Interleukin-2/metabolism , Dendritic Cells/metabolism , Cell Proliferation
2.
Clin Cancer Res ; 28(10): 2082-2093, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35262675

ABSTRACT

PURPOSE: IL15 promotes activation and maintenance of natural killer (NK) and CD8+ T effector memory cells making it a potential immunotherapeutic agent for the treatment of cancer. However, monotherapy with IL15 was ineffective in patients with cancer, indicating that it would have to be used in combination with other anticancer agents. The administration of high doses of common gamma chain cytokines, such as IL15, is associated with the generation of "helpless" antigen-nonspecific CD8 T cells. The generation of the tumor-specific cytotoxic T cells can be mediated by CD40 signaling via agonistic anti-CD40 antibodies. Nevertheless, parenteral administration of anti-CD40 antibodies is associated with unacceptable side effects, such as thrombocytopenia and hepatic toxicity, which can be avoided by intratumoral administration. EXPERIMENTAL DESIGN: We investigated the combination of IL15 with an intratumoral anti-CD40 monoclonal antibody (mAb) in a dual tumor TRAMP-C2 murine prostate cancer model and expanded the regimen to include an anti-PD-1 mAb. RESULTS: Here we demonstrated that anti-CD40 given intratumorally not only showed significant antitumor activity in treated tumors, but also noninjected contralateral tumors, indicative of abscopal efficacy. The combination of IL15 with intratumoral anti-CD40 showed an additive immune response with an increase in the number of tumor-specific tetramer-positive CD8 T cells. Furthermore, the addition of anti-PD-1 further improved efficacy mediated by the anti-CD40/IL15 combination. CONCLUSIONS: These studies support the initiation of a clinical trial in patients with cancer using IL15 in association with the checkpoint inhibitor, anti-PD-1, and intratumoral optimized anti-CD40.


Subject(s)
Antineoplastic Agents , Neoplasms , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/pharmacology , CD40 Antigens , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Disease Models, Animal , Humans , Interleukin-15 , Male , Mice , Mice, Inbred C57BL
3.
Blood Adv ; 6(7): 2346-2360, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35030628

ABSTRACT

Adult T-cell leukemia/lymphoma (ATL) is an aggressive T-cell lymphoproliferative malignancy caused by human T-cell leukemia virus type 1 (HTLV-1). ATL is an orphan disease with no curative drug treatment regimens urgently needing new combination therapy. HTLV-1-infected cells rely on viral proteins, Tax and HBZ (HTLV-1-b-ZIP factor), to activate the transcription of various host genes that are critical for promoting leukemic transformation. Inhibition of bromodomain and extraterminal motif (BET) protein was previously shown to collapse the transcriptional network directed by BATF3 super-enhancer and thereby induced ATL cell apoptosis. In the current work, by using xenograft, ex vivo, and in vitro models, we demonstrated that I-BET762 (BETi) synergized with copanlisib (PI3Ki) and bardoxolone methyl (NF-κBi) to dramatically decrease the growth of ATL cells. Mechanistically, the triple combination exhibited synergistic activity by down-regulating the expression of c-MYC while upregulating the level of the glucocorticoid-induced leucine zipper (GILZ). The triple combination also enhanced apoptosis induction by elevating the expression of active caspase-3 and cleaved PARP. Importantly, the triple combination prolonged the survival of ATL-bearing xenograft mice and inhibited the proliferation of ATL cells from peripheral blood mononuclear cells (PBMCs) of both acute and smoldering/chronic ATL patients. Therefore, our data provide the rationale for a clinical trial exploring the multiagent combination of BET, PI3K/AKT, and NF-κB inhibitors for ATL patients and expands the potential treatments for this recalcitrant malignancy.


Subject(s)
Human T-lymphotropic virus 1 , Leukemia-Lymphoma, Adult T-Cell , Lymphoma , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Human T-lymphotropic virus 1/genetics , Humans , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/pathology , Leukocytes, Mononuclear/metabolism , Mice , NF-kappa B/metabolism , Oleanolic Acid/analogs & derivatives , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/therapeutic use
4.
Transl Oncol ; 14(1): 100913, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33129109

ABSTRACT

Adult T-cell leukemia (ATL) is an aggressive T-cell lymphoproliferative malignancy of regulatory T lymphocytes (Tregs), caused by human T-cell lymphotropic virus 1 (HTLV-1). Interleukin 2 receptor alpha (IL-2Rα) is expressed in the leukemic cells of smoldering/chronic ATL patients, leading to constitutive activation of the JAK/STAT pathway and spontaneous proliferation. The PI3K/AKT/mTOR pathway also plays a critical role in ATL cell survival and proliferation. We previously performed a high-throughput screen that demonstrated additive/synergistic activity of Ruxolitinib, a JAK1/2 inhibitor, with AZD8055, an mTORC1/C2 inhibitor. However, effects of unintended JAK2 inhibition with Ruxolitinib limits it therapeutic potential for ATL patients, which lead us to evaluate a JAK1-specific inhibitor. Here, we demonstrated that Upadacitinib, a JAK-1 inhibitor, inhibited the proliferation of cytokine-dependent ATL cell lines and the expression of p-STAT5. Combinations of Upadacitinib with either AZD8055 or Sapanisertib, mTORC1/C2 inhibitors, showed anti-proliferative effects against cytokine-dependent ATL cell lines and synergistic effect with reducing tumor growth in NSG mice bearing IL-2 transgenic tumors. Importantly, the combination of these two agents inhibited ex vivo spontaneous proliferation of ATL cells from patients with smoldering/chronic ATL. Combined targeting of JAK/STAT and PI3K/AKT/mTOR pathways represents a promising therapeutic intervention for patients with smoldering/chronic ATL.

5.
Proc Natl Acad Sci U S A ; 115(46): E10915-E10924, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30373815

ABSTRACT

The goal of cancer immunotherapy is to stimulate the host immune system to attack malignant cells. Antibody-dependent cellular cytotoxicity (ADCC) is a pivotal mechanism of antitumor action of clinically employed antitumor antibodies. IL-15 administered to patients with metastatic malignancy by continuous i.v. infusion at 2 µg/kg/d for 10 days was associated with a 38-fold increase in the number and activation status of circulating natural killer (NK) cells and activation of macrophages which together are ADCC effectors. We investigated combination therapy of IL-15 with rituximab in a syngeneic mouse model of lymphoma transfected with human CD20 and with alemtuzumab (Campath-1H) in a xenograft model of human adult T cell leukemia (ATL). IL-15 greatly enhanced the therapeutic efficacy of both rituximab and alemtuzumab in tumor models. The additivity/synergy was shown to be associated with augmented ADCC. Both NK cells and macrophages were critical elements in the chain of interacting effectors involved in optimal therapeutic responses mediated by rituximab with IL-15. We provide evidence supporting the hypothesis that NK cells interact with macrophages to augment the NK-cell activation and expression of FcγRIV and the capacity of these cells to become effectors of ADCC. The present study supports clinical trials of IL-15 combined with tumor-directed monoclonal antibodies.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Interleukin-15/administration & dosage , Killer Cells, Natural/immunology , Macrophages/immunology , Alemtuzumab/administration & dosage , Animals , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity/drug effects , Cell Line, Tumor , Female , Humans , Interleukin-15/immunology , Killer Cells, Natural/drug effects , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Rituximab/administration & dosage
6.
Proc Natl Acad Sci U S A ; 114(15): 3975-3980, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28356514

ABSTRACT

Activating Janus kinase (JAK) and signal transducer and activator of transcription (STAT) mutations have been discovered in many T-cell malignancies, including anaplastic lymphoma kinase (ALK)- anaplastic large cell lymphomas (ALCLs). However, such mutations occur in a minority of patients. To investigate the clinical application of targeting JAK for ALK- ALCL, we treated ALK- cell lines of various histological origins with JAK inhibitors. Interestingly, most exogenous cytokine-independent cell lines responded to JAK inhibition regardless of JAK mutation status. JAK inhibitor sensitivity correlated with the STAT3 phosphorylation status of tumor cells. Using retroviral shRNA knockdown, we have demonstrated that these JAK inhibitor-sensitive cells are dependent on both JAK1 and STAT3 for survival. JAK1 and STAT3 gain-of-function mutations were found in some, but not all, JAK inhibitor-sensitive cells. Moreover, the mutations alone cannot explain the JAK1/STAT3 dependency, given that wild-type JAK1 or STAT3 was sufficient to promote cell survival in the cells that had either JAK1or STAT3 mutations. To investigate whether other mechanisms were involved, we knocked down upstream receptors GP130 or IL-2Rγ. Knockdown of GP130 or IL-2Rγ induced cell death in selected JAK inhibitor-sensitive cells. High expression levels of cytokines, including IL-6, were demonstrated in cell lines as well as in primary ALK- ALCL tumors. Finally, ruxolitinib, a JAK1/2 inhibitor, was effective in vivo in a xenograft ALK- ALCL model. Our data suggest that cytokine receptor signaling is required for tumor cell survival in diverse forms of ALK- ALCL, even in the presence of JAK1/STAT3 mutations. Therefore, JAK inhibitor therapy might benefit patients with ALK- ALCL who are phosphorylated STAT3.


Subject(s)
Janus Kinase 1/genetics , Lymphoma, Large-Cell, Anaplastic/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cytokine/metabolism , STAT3 Transcription Factor/genetics , Anaplastic Lymphoma Kinase , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Cytokines/genetics , Cytokines/metabolism , Humans , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Lymphoma, Large-Cell, Anaplastic/drug therapy , Lymphoma, Large-Cell, Anaplastic/genetics , Mice , Nitriles , Phosphorylation , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Cytokine/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
7.
Proc Natl Acad Sci U S A ; 113(6): 1624-9, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26811457

ABSTRACT

Despite relative success of therapy for Hodgkin's lymphoma (HL), novel therapeutic agents are needed for patients with refractory or relapsed disease. Recently, anti-PD1 immunotherapy or treatment with the anti-CD30 toxin conjugate brentuximab vedotin (BV) have been associated with remissions; however, the median responses of complete responses (CRs) with the latter were only 6.7 mo. To obtain curative therapy, other effective agents, based on HL biology, would have to be given in combination with BV. Hodgkin's Reed-Sternberg (HRS) cells secrete cytokines including IL-6 and -13, leading to constitutive activation of JAK/STAT signaling. In the present study the JAK1/2 inhibitor ruxolitinib reduced phosphorylation of STAT3 and STAT6 and expression of c-Myc in the HL cell line HDLM-2. These changes were enhanced when, on the basis of a matrix screen of drug combinations, ruxolitinib was combined with the Bcl-2/Bcl-xL inhibitor Navitoclax. The combination augmented expression of Bik, Puma, and Bax, and attenuated Bcl-xL expression and the phosphorylation of Bad. The use of the two-agent combination of either ruxolitinib or Navitoclax with BV or the three-agent combination strongly activated Bax and increased activities of cytochrome c and caspase-9 and -3 that, in turn, led to cleavage of poly(ADP ribose) polymerase and Mcl-1. Either ruxolitinib combined with Navitoclax or BV alone prolonged survival but did not cure HDLM-2 tumor-bearing mice, whereas BV combined with ruxolitinib and/or with Navitoclax resulted in a sustained, complete elimination of the HDLM-2 HL. These studies provide scientific support for a clinical trial to evaluate BV combined with ruxolitinib in select patients with HL.


Subject(s)
Aniline Compounds/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hodgkin Disease/drug therapy , Immunoconjugates/therapeutic use , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use , Aniline Compounds/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Blotting, Western , Brentuximab Vedotin , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Disease Progression , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Gene Dosage , Hodgkin Disease/enzymology , Hodgkin Disease/pathology , Humans , Immunoconjugates/pharmacology , Janus Kinase 2/genetics , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Nitriles , Phosphorylation/drug effects , Pyrazoles/pharmacology , Pyrimidines , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Sulfonamides/pharmacology , Treatment Outcome , bcl-X Protein/metabolism
8.
Proc Natl Acad Sci U S A ; 112(40): 12480-5, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26396258

ABSTRACT

Adult T-cell leukemia (ATL) develops in individuals infected with human T-cell lymphotropic virus-1 (HTLV-1). Presently there is no curative therapy for ATL. HTLV-1-encoded protein Tax (transactivator from the X-gene region) up-regulates Bcl-xL (B-cell lymphoma-extra large) expression and activates interleukin-2 (IL-2), IL-9, and IL-15 autocrine/paracrine systems, resulting in amplified JAK/STAT signaling. Inhibition of JAK signaling reduces cytokine-dependent ex vivo proliferation of peripheral blood mononuclear cells (PBMCs) from ATL patients in smoldering/chronic stages. Currently, two JAK inhibitors are approved for human use. In this study, we examined activity of multiple JAK inhibitors in ATL cell lines. The selective JAK inhibitor ruxolitinib was examined in a high-throughput matrix screen combined with >450 potential therapeutic agents, and Bcl-2/Bcl-xL inhibitor navitoclax was identified as a strong candidate for multicomponent therapy. The combination was noted to strongly activate BAX (Bcl-2-associated X protein), effect mitochondrial depolarization, and increase caspase 3/7 activities that lead to cleavage of PARP (poly ADP ribose polymerase) and Mcl-1 (myeloid cell leukemia 1). Ruxolitinib and navitoclax independently demonstrated modest antitumor efficacy, whereas the combination dramatically lowered tumor burden and prolonged survival in an ATL murine model. This combination strongly blocked ex vivo proliferation of five ATL patients' PBMCs. These studies provide support for a therapeutic trial in patients with smoldering/chronic ATL using a drug combination that inhibits JAK signaling and antiapoptotic protein Bcl-xL.


Subject(s)
Interleukin-2/metabolism , Janus Kinases/metabolism , Leukemia-Lymphoma, Adult T-Cell/metabolism , STAT Transcription Factors/metabolism , bcl-X Protein/metabolism , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Female , Flow Cytometry , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Janus Kinases/antagonists & inhibitors , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/pathology , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Nitriles , Pyrazoles/pharmacology , Pyrimidines , STAT Transcription Factors/antagonists & inhibitors , Signal Transduction/drug effects , Sulfonamides/pharmacology , Xenograft Model Antitumor Assays , bcl-X Protein/antagonists & inhibitors
9.
Eur J Immunol ; 44(11): 3330-41, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25142300

ABSTRACT

Interleukin-15 (IL-15) is an inflammatory cytokine whose role in autoimmune diseases has not been fully elucidated. Th17 cells have been shown to play critical roles in experimental autoimmune encephalomyelitis (EAE) models. In this study, we demonstrate that blockade of IL-15 signaling by TMß-1 mAb treatment aggravated EAE severity. The key mechanism was not NK-cell depletion but depletion of CD8+ CD122+ T cells. Adoptive transfer of exogenous CD8+ CD122+ T cells to TMß-1-treated mice rescued animals from severe disease. Moreover, transfer of preactivated CD8+ CD122+ T cells prevented EAE development and significantly reduced IL-17 secretion. Naïve effector CD4+ CD25- T cells cultured with either CD8+ CD122+ T cells from wild-type mice or IL-15 transgenic mice displayed lower frequencies of IL-17A production with lower amounts of IL-17 in the supernatants when compared with production by effector CD4+ CD25- T cells cultured alone. Addition of a neutralizing antibody to IL-10 led to recovery of IL-17A production in Th17 cultures. Furthermore, coculture of CD8+ CD122+ T cells with effector CD4+ T cells inhibited their proliferation significantly, suggesting a regulatory function for IL-15 dependent CD8+ CD122+ T cells. Taken together, these observations suggest that IL-15, acting through CD8+ CD122+ T cells, has a negative regulatory role in reducing IL-17 production and Th17-mediated EAE inflammation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-15/immunology , Interleukin-17/biosynthesis , Th17 Cells/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Interleukin-15/antagonists & inhibitors , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-2 Receptor beta Subunit/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Lymphocyte Activation/immunology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction/immunology , Th17 Cells/cytology
10.
J Innate Immun ; 6(3): 377-93, 2014.
Article in English | MEDLINE | ID: mdl-24577058

ABSTRACT

Toll/IL-1R domain-containing adaptor inducing interferon-ß (IFN-ß) factor (TRIF) is a key adaptor for Toll-like receptor (TLR) 3 and TLR4 signaling. Using a novel cDNA isolate encoding a TRIF protein with a 21-residue deletion (Δ160-181) from its amino-terminal half, we investigated the impact of this deletion on TRIF functions. Transfection studies consistently showed higher expression levels of the (Δ160-181) TRIF compared to wild-type (wt) TRIF, an effect unrelated to apoptosis, cell lines or plasmid amplification. Colocalization of wt and (Δ160-181) TRIF proteins led to a dramatic reduction of their respective expressions, suggesting that wt/(Δ160-181) TRIF heterocomplexes are targeted for degradation. We demonstrated that wt TRIF associates with tumor necrosis factor-α receptor-associated factor 3 (TRAF3) better than (Δ160-181) TRIF, culminating in its greater ubiquitination and proteolysis. This explains, in part, the differential expression levels of the two TRIF proteins. Despite higher expression levels in transfected cells, (Δ160-181) TRIF inefficiently transactivated the IFN pathway, whereas the nuclear factor-κB (NF-κB) pathway activation remained similar to that by wt TRIF. In coexpression studies, (Δ160-181) TRIF marginally contributed to the IFN pathway activation, but still enhanced NF-κB signaling with wt TRIF. Therefore, this 21 amino acid sequence is crucial for TRAF3 association, modulation of TRIF stability and activation of the IFN pathway.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Interferons/metabolism , TNF Receptor-Associated Factor 3/metabolism , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Vesicular Transport/genetics , HeLa Cells , Humans , NF-kappa B/metabolism , Protein Binding/genetics , Protein Stability , Protein Structure, Tertiary/genetics , Protein Transport , Proteolysis , Sequence Deletion/genetics , Signal Transduction/genetics , Transcriptional Activation/genetics
11.
Blood ; 117(15): 4032-40, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21304101

ABSTRACT

IL-15 has growth-promoting effects on select lymphoid subsets, including natural killer (NK) cells, NK T cells, intraepithelial lymphocytes (IELs), CD8 T cells, and γδ-T cells. Constitutive expression of murine IL-15 in IL-15-transgenic mice was reported to cause T-NK leukemia. We investigated whether IL-15 expression is sufficient for leukemic transformation using a human IL-15-transgenic (IL-15Tg) mouse model. We noted that 100% of the mice observed over a 2-year period (n > 150) developed fatal expansions of CD8 T cells with NK markers, and determined that these cells expressed IL-15 receptor alpha (IL-15Rα). The expression of IL-15Rα on CD8 T cells appears to be required for uncontrolled aggressive lymphoproliferation, because none of the IL-15Rα(-/-)-IL-15Tg mice that we followed for more than 2 years developed the fatal disease despite controlled expansion of CD8 T cells. In addition, in contrast to IL-15Tg mice, in which leukemia-like CD8 T cells expressed IL-15Rα persistently, acutely activated normal CD8 T cells only transiently expressed IL-15Rα. Inhibition of DNA methylation enabled sustained IL-15Rα expression induced by activation. We present a scenario for IL-15Tg mice in which CD8 T cells that acquire constitutive persistent IL-15Rα expression are at a selective advantage and become founder cells, outgrow other lymphocytes, and lead to the establishment of a leukemia-like condition.


Subject(s)
Autocrine Communication/immunology , CD8-Positive T-Lymphocytes/physiology , Interleukin-15 Receptor alpha Subunit/genetics , Interleukin-15/genetics , Leukemia, T-Cell , Animals , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Epigenesis, Genetic/physiology , Gene Silencing/physiology , Genetic Complementation Test , Homeostasis/immunology , Humans , Interleukin-15/metabolism , Interleukin-15 Receptor alpha Subunit/metabolism , Leukemia, T-Cell/genetics , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation
12.
Hum Genet ; 110(5): 422-8, 2002 May.
Article in English | MEDLINE | ID: mdl-12073012

ABSTRACT

TDGF1 (CRIPTO) is an EGF-CFC family member and an obligate co-receptor involved in NODAL signaling, a developmental program implicated in midline, forebrain, and left-right axis development in model organisms. Previous studies of CFC1 (CRYPTIC), another member of the EGF-CFC family, demonstrated that normal function of this protein is required for proper laterality development in humans. Here we identify a mutation in the conserved CFC domain of TDGF1 in a patient with midline anomalies of the forebrain. The mutant protein is inactive in a zebrafish rescue assay, indicating a role for TDGF1 in human midline and forebrain development.


Subject(s)
Epidermal Growth Factor , Holoprosencephaly/genetics , Homeodomain Proteins , Membrane Glycoproteins , Mutation/genetics , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Prosencephalon/abnormalities , Transcription Factors , Zebrafish Proteins , Amino Acid Motifs , Amino Acid Sequence , Animals , Child, Preschool , Female , GPI-Linked Proteins , Humans , Intercellular Signaling Peptides and Proteins , Male , Molecular Sequence Data , Polymerase Chain Reaction , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Zebrafish/embryology , Zebrafish/genetics
SELECTION OF CITATIONS
SEARCH DETAIL