Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 211
Filter
1.
Adv Immunol ; 131: 187-231, 2016.
Article in English | MEDLINE | ID: mdl-27235684

ABSTRACT

Positron emission tomography (PET) is a powerful noninvasive imaging technique able to measure distinct biological processes in vivo by administration of a radiolabeled probe. Whole-body measurements track the probe accumulation providing a means to measure biological changes such as metabolism, cell location, or tumor burden. PET can also be applied to both preclinical and clinical studies providing three-dimensional information. For immunotherapies (in particular understanding T cell responses), PET can be utilized for spatial and longitudinal tracking of T lymphocytes. Although PET has been utilized clinically for over 30 years, the recent development of additional PET radiotracers have dramatically expanded the use of PET to detect endogenous or adoptively transferred T cells in vivo. Novel probes have identified changes in T cell quantity, location, and function. This has enabled investigators to track T cells outside of the circulation and in hematopoietic organs such as spleen, lymph nodes, and bone marrow, or within tumors. In this review, we cover advances in PET detection of the antitumor T cell response and areas of focus for future studies.


Subject(s)
Immunotherapy, Adoptive/methods , Neoplasms/therapy , Positron-Emission Tomography/methods , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/immunology , Cell Movement , Humans , Imaging, Three-Dimensional , Lymphocyte Activation , Molecular Imaging , Neoplasms/immunology , Positron-Emission Tomography/trends , T-Lymphocytes/transplantation
2.
Leukemia ; 25(2): 290-300, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21102429

ABSTRACT

ABL gene translocations create constitutively active tyrosine kinases that are causative in chronic myeloid leukemia, acute lymphocytic leukemia and other hematopoietic malignancies. Consistent retention of ABL SH3/SH2 autoinhibitory domains, however, suggests that these leukemogenic tyrosine kinase fusion proteins remain subject to regulation. We resolve this paradox, demonstrating that BCR-ABL1 kinase activity is regulated by RIN1, an ABL SH3/SH2 binding protein. BCR-ABL1 activity was increased by RIN1 overexpression and decreased by RIN1 silencing. Moreover, Rin1(-/-) bone marrow cells were not transformed by BCR-ABL1, ETV6-ABL1 or BCR-ABL1(T315I), a patient-derived drug-resistant mutant, as judged by growth factor independence. Rescue by ectopic RIN1 verified a cell autonomous mechanism of collaboration with BCR-ABL1 during transformation. Sensitivity to the ABL kinase inhibitor imatinib was increased by RIN1 silencing, consistent with RIN1 stabilization of an activated BCR-ABL1 conformation having reduced drug affinity. The dependence on activation by RIN1 to unleash full catalytic and cell transformation potential reveals a previously unknown vulnerability that could be exploited for treatment of leukemic cases driven by ABL translocations. The findings suggest that RIN1 targeting could be efficacious for imatinib-resistant disease and might complement ABL kinase inhibitors in first-line therapy.


Subject(s)
Cell Transformation, Neoplastic , Fusion Proteins, bcr-abl , Genes, abl , Intracellular Signaling Peptides and Proteins/physiology , Protein Kinase Inhibitors/pharmacology , Animals , Benzamides , Humans , Imatinib Mesylate , K562 Cells , Mice , Piperazines/pharmacology , Pyrimidines/pharmacology , Translocation, Genetic , src Homology Domains
3.
Article in English | MEDLINE | ID: mdl-19022743

ABSTRACT

Prostate cancer is a leading cause of cancer-related death in adult men. It can regress dramatically upon antihormonal therapy, but it often recurs in a more aggressive, androgen-independent form. Defining the prostate tissue stem cells (PrSCs) and their involvement in cancer initiation and maintenance may lead to better therapeutics. Using a tissue-regeneration model in which dissociated prostate epithelial cells mixed with inductive mesenchyme give rise to prostatic tubules, we have identified a small population of prostate cells that contains multiple stem cell characteristics. In this system, prostate cancer can be initiated by autocrine or paracrine growth factor signaling and intracellular overexpression of genes often found mutated in human prostate cancer. Using an in vitro prostate sphere assay, we further defined the PrSC population and demonstrated their self-renewal and multilineage differentiation capabilities. Microarray analyses of the stem- and non-stem-cell populations have assisted us in finding and evaluating additional markers that can better define the PrSC population and further delineate the different cell types of the prostate, including those that serve as the target cell for tumor initiation.


Subject(s)
Adult Stem Cells/cytology , Neoplastic Stem Cells/pathology , Prostate/cytology , Prostatic Neoplasms/pathology , Adult , Adult Stem Cells/metabolism , Animals , Cell Culture Techniques , Cell Differentiation , Colony-Forming Units Assay , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Male , Mice , Models, Biological , Neoplastic Stem Cells/metabolism , Prostate/metabolism , Prostatic Neoplasms/etiology , Prostatic Neoplasms/genetics , Regeneration
4.
Article in English | MEDLINE | ID: mdl-16869753

ABSTRACT

Understanding prostate stem cells (PSCs) may provide insight for the design of therapeutics for prostate cancer. We have developed a quantitative in vivo colony-forming assay and have demonstrated that the Sca-1 antigen is present on the surface of a prostate cell subpopulation that possesses multiple stem cell properties. Immunofluorescent analysis demonstrates that Sca-1 is expressed by both basal and luminal cells in the proximal region of the adult prostate, but is not expressed by either lineage in more distal regions. The proximal region has been suggested as the PSC niche based on BrdU label-retention studies and the presence of distinct smooth-muscle cells that produce high levels of TGF-beta. Sca-1 is also expressed by nearly all cells within fetal prostate epithelial chords, suggesting Sca-1 may be conserved on PSCs throughout development. Malignant epithelial cells from TRAMP mice, as well as normal prostate cells with lentiviral-mediated alteration of the PTEN/AKT signaling pathway, give rise to PIN lesions and prostate cancer in vivo. Alteration of PTEN/AKT signaling in Sca-1-enriched PSCs also results in PIN lesions, suggesting that PSCs can serve as one target for prostate carcinogenesis.


Subject(s)
Neoplastic Stem Cells/pathology , Prostate/cytology , Prostatic Neoplasms/pathology , Stem Cells/cytology , Animals , Antigens, Ly/metabolism , Colony-Forming Units Assay , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Neoplastic Stem Cells/metabolism , PTEN Phosphohydrolase/metabolism , Prostate/metabolism , Prostatic Neoplasms/etiology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/therapy , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Stem Cells/metabolism , Transforming Growth Factor beta/metabolism , Tumor Stem Cell Assay
5.
Proc Natl Acad Sci U S A ; 101(50): 17456-61, 2004 Dec 14.
Article in English | MEDLINE | ID: mdl-15505216

ABSTRACT

Protein kinase inhibitors can be effective in treating selected cancers, but most suppress several kinases. Imatinib mesylate has been useful in the treatment of Philadelphia chromosome-positive chronic myelogenous leukemia and B cell acute lymphoblastic leukemia through the inhibition of BCR-ABL tyrosine kinase activity. Imatinib mesylate has also been shown to inhibit KIT, ARG, and platelet-derived growth factor receptors alpha and beta, and potentially other tyrosine kinases. We have produced a mutant allele of BCR-ABL (T315A) that is uniquely inhibitable by the small molecule 4-amino-1-tert-butyl-3-(1-naphthyl)pyrazolo[3,4-d]pyrimidine and used it to demonstrate that sole suppression of BCR-ABL activity was insufficient to eliminate BCR-ABL(+) KIT(+)-expressing immature murine myeloid leukemic cells. In contrast, imatinib mesylate effectively eliminated BCR-ABL(+) KIT(+)-expressing leukemic cells. In the cellular context of mature myeloid cells and Pro/Pre B cells that do not express KIT, monospecific BCR-ABL inhibition was quantitatively as effective as imatinib mesylate in suppressing cell growth and inducing apoptosis. These results suggest that the therapeutic effectiveness of small molecule drugs such as imatinib mesylate could be due to the inhibitor's ability to suppress protein kinases in addition to the dominant target.


Subject(s)
Enzyme Inhibitors/pharmacology , Myeloproliferative Disorders/enzymology , Myeloproliferative Disorders/pathology , Phosphotyrosine , Protein-Tyrosine Kinases/antagonists & inhibitors , Apoptosis/drug effects , B-Lymphocytes/drug effects , B-Lymphocytes/pathology , Benzamides , Cell Cycle/drug effects , Cell Line , Cell Proliferation/drug effects , Drug Synergism , Fusion Proteins, bcr-abl , Gene Expression Regulation, Enzymologic , Humans , Imatinib Mesylate , Mutation/genetics , Myeloproliferative Disorders/genetics , Phosphotyrosine/metabolism , Piperazines/pharmacology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Pyrimidines/pharmacology , Tyrosine/genetics , Tyrosine/metabolism
7.
EMBO J ; 20(20): 5692-702, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11598012

ABSTRACT

Mutations in Bruton's tyrosine kinase (Btk) result in X-linked agammaglobulinemia (XLA) in humans and X-linked immunodeficiency (xid) in mice. While targeted disruption of the protein kinase C-beta (PKCbeta) gene in mice results in an immunodeficiency similar to xid, the overall tyrosine phosphorylation of Btk is significantly enhanced in PKCbeta-deficient B cells. We provide direct evidence that PKCbeta acts as a feedback loop inhibitor of Btk activation. Inhibition of PKCbeta results in a dramatic increase in B-cell receptor (BCR)-mediated Ca2+ signaling. We identified a highly conserved PKCbeta serine phosphorylation site in a short linker within the Tec homology domain of Btk. Mutation of this phosphorylation site led to enhanced tyrosine phosphorylation and membrane association of Btk, and augmented BCR and FcepsilonRI-mediated signaling in B and mast cells, respectively. These findings provide a novel mechanism whereby reversible translocation of Btk/Tec kinases regulates the threshold for immunoreceptor signaling and thereby modulates lymphocyte activation.


Subject(s)
Isoenzymes/physiology , Lymphocyte Activation/physiology , Membrane Proteins/metabolism , Protein Kinase C/physiology , Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, B-Cell/physiology , 3T3 Cells , Agammaglobulinaemia Tyrosine Kinase , Agammaglobulinemia/genetics , Alleles , Amino Acid Sequence , Animals , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , Calcium Signaling/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Feedback , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/deficiency , Isoenzymes/genetics , Mast Cells/enzymology , Mast Cells/immunology , Mice , Mice, Knockout , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Mapping , Phosphorylation , Phosphoserine/chemistry , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/deficiency , Protein Kinase C/genetics , Protein Kinase C beta , Protein Processing, Post-Translational , Protein Structure, Tertiary , Protein Transport , Protein-Tyrosine Kinases/chemistry , Receptors, IgE/physiology
8.
Oncogene ; 20(40): 5644-59, 2001 Sep 10.
Article in English | MEDLINE | ID: mdl-11607816

ABSTRACT

The Ph chromosome has been genetically linked to CML and ALL. Its chimeric fusion gene product, BCR-ABL, can generate leukemia in mice. This review will discuss selected model systems developed to study BCR-ABL induced leukemia and focuses on what we have learned about the human disease from these models. Five main experimental approaches will be discussed including: (i) Reconstitution of mice with bone marrow cells retrovirally transduced with BCR-ABL; (ii) Transgenic mice expressing BCR-ABL; (iii) Knock-in mice with BCR-ABL expression driven from the endogenous bcr locus; (iv) Development of CML-like disease in mice with loss of function mutations in heterologous genes; and (v) ES in vitro hematopoietic differentiation coupled with regulated BCR-ABL expression.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology , Animals , Bone Marrow Cells/metabolism , Cell Differentiation , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Genetic Linkage , Humans , Mice , Mice, Knockout , Mice, Transgenic , Models, Genetic , Oncogene Proteins, Fusion/metabolism , Phenotype , Promoter Regions, Genetic , Protein Structure, Tertiary , Retroviridae/genetics , Transduction, Genetic
9.
J Biol Chem ; 276(44): 41325-35, 2001 Nov 02.
Article in English | MEDLINE | ID: mdl-11535583

ABSTRACT

Sphingosylphosphorylcholine (SPC) and lysophosphatidylcholine (LPC) are bioactive lipid molecules involved in numerous biological processes. We have recently identified ovarian cancer G protein-coupled receptor 1 (OGR1) as a specific and high affinity receptor for SPC, and G2A as a receptor with high affinity for LPC, but low affinity for SPC. Among G protein-coupled receptors, GPR4 shares highest sequence homology with OGR1 (51%). In this work, we have identified GPR4 as not only another high affinity receptor for SPC, but also a receptor for LPC, albeit of lower affinity. Both SPC and LPC induce increases in intracellular calcium concentration in GPR4-, but not vector-transfected MCF10A cells. These effects are insensitive to treatment with BN52021, WEB-2170, and WEB-2086 (specific platelet activating factor (PAF) receptor antagonists), suggesting that they are not mediated through an endogenous PAF receptor. SPC and LPC bind to GPR4 in GPR4-transfected CHO cells with K(d)/SPC = 36 nm, and K(d)/LPC = 159 nm, respectively. Competitive binding is elicited only by SPC and LPC. Both SPC and LPC activate GPR4-dependent activation of serum response element reporter and receptor internalization. Swiss 3T3 cells expressing GPR4 respond to both SPC and LPC, but not sphingosine 1-phosphate (S1P), PAF, psychosine (Psy), glucosyl-beta1'1-sphingosine (Glu-Sph), galactosyl-beta1'1-ceramide (Gal-Cer), or lactosyl-beta1'1-ceramide (Lac-Cer) to activate extracellular signal-regulated kinase mitogen-activated protein kinase in a concentration- and time-dependent manner. SPC and LPC stimulate DNA synthesis in GPR4-expressing Swiss 3T3 cells. Both extracellular signal-regulated kinase activation and DNA synthesis stimulated by SPC and LPC are pertussis toxin-sensitive, suggesting the involvement of a G(i)-heterotrimeric G protein. In addition, GPR4 expression confers chemotactic responses to both SPC and LPC in Swiss 3T3 cells. Taken together, our data indicate that GPR4 is a receptor with high affinity to SPC and low affinity to LPC, and that multiple cellular functions can be transduced via this receptor.


Subject(s)
Lysophosphatidylcholines/metabolism , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , Sphingosine/analogs & derivatives , Sphingosine/metabolism , 3T3 Cells , Animals , Base Sequence , CHO Cells , Calcium/metabolism , Cricetinae , DNA Primers , DNA Replication , Enzyme Activation , Humans , Ligands , Mice , Mitogen-Activated Protein Kinases/metabolism , Radioligand Assay , Receptors, Cell Surface/genetics , Transfection
10.
Science ; 293(5530): 702-5, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11474113

ABSTRACT

Although the biological actions of the cell membrane and serum lipid lysophosphatidylcholine (LPC) in atherosclerosis and systemic autoimmune disease are well recognized, LPC has not been linked to a specific cell-surface receptor. We show that LPC is a high-affinity ligand for G2A, a lymphocyte-expressed G protein-coupled receptor whose genetic ablation results in the development of autoimmunity. Activation of G2A by LPC increased intracellular calcium concentration, induced receptor internalization, activated ERK mitogen-activated protein kinase, and modified migratory responses of Jurkat T lymphocytes. This finding implicates a role for LPC-G2A interaction in the etiology of inflammatory autoimmune disease and atherosclerosis.


Subject(s)
Cell Cycle Proteins/metabolism , Lysophosphatidylcholines/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , T-Lymphocytes/metabolism , Animals , Arteriosclerosis/immunology , Arteriosclerosis/metabolism , Calcium/metabolism , Cell Line , Chemotaxis, Leukocyte , Humans , Jurkat Cells , Ligands , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lymphocyte Activation , Lysophosphatidylcholines/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/metabolism , Phosphorylcholine/pharmacology , Radioligand Assay , Recombinant Fusion Proteins/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine/pharmacology , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Virulence Factors, Bordetella/pharmacology
11.
BMC Immunol ; 2: 4, 2001.
Article in English | MEDLINE | ID: mdl-11410123

ABSTRACT

BACKGROUND: Bruton's tyrosine kinase (Btk) is essential for B cell development and function. Mutations of Btk elicit X-linked agammaglobulinemia in humans and X-linked immunodeficiency in the mouse. Btk has been proposed to participate in B cell antigen receptor-induced signaling events leading to activation of phospholipase C-gamma2 (PLCgamma2) and calcium mobilization. However it is unclear whether Btk activation is alone sufficient for these signaling events, and whether Btk can activate additional pathways that do not involve PLCgamma2. To address such issues we have generated Btk:ER, a conditionally active form of the kinase, and expressed it in the PLCgamma2-deficient DT40 B cell line. RESULTS: Activation of Btk:ER was sufficient to induce multiple B cell signaling pathways in PLCgamma2-sufficient DT40 cells. These included tyrosine phosphorylation of PLCgamma2, mobilization of intracellular calcium, activation of extracellular signal-regulated kinase (ERK) and c-Jun NH2-terminal kinase (JNK) mitogen-activated protein kinase (MAPK) pathways, and apoptosis. In DT40 B cells deficient for PLCgamma2, Btk:ER activation failed to induce the signaling events described above with the consequence that the cells failed to undergo apoptosis. CONCLUSIONS: These data suggest that Btk:ER regulates downstream signaling pathways primarily via PLCgamma2 in B cells. While it is not known whether activated Btk:ER precisely mimics activated Btk, this conditional system will likely facilitate the dissection of the role of Btk and its family members in a variety of biological processes in many different cell types.


Subject(s)
B-Lymphocytes/enzymology , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Type C Phospholipases/physiology , Agammaglobulinaemia Tyrosine Kinase , Animals , Apoptosis , B-Lymphocytes/immunology , Calcium Signaling , Cell Line , MAP Kinase Signaling System , Mice , Mutation , Phospholipase C gamma , Protein-Tyrosine Kinases/genetics , Receptors, Estrogen/genetics , Recombinant Fusion Proteins/metabolism , Type C Phospholipases/genetics
12.
Immunity ; 14(5): 561-71, 2001 May.
Article in English | MEDLINE | ID: mdl-11371358

ABSTRACT

Mice with a targeted disruption of the gene encoding a lymphoid-expressed orphan G protein-coupled receptor, G2A, demonstrate a normal pattern of T and B lineage differentiation through young adulthood. As G2A-deficient animals age, they develop secondary lymphoid organ enlargement associated with abnormal expansion of both T and B lymphocytes. Older G2A-deficient mice (>1 year) develop a slowly progressive wasting syndrome, characterized by lymphocytic infiltration into various tissues, glomerular immune complex deposition, and anti-nuclear autoantibodies. G2A-deficient T cells are hyperresponsive to TCR stimulation, exhibiting enhanced proliferation and a lower threshold for activation. Our findings demonstrate that G2A plays a critical role in controlling peripheral lymphocyte homeostasis and that its ablation results in the development of a novel, late-onset autoimmune syndrome.


Subject(s)
Autoimmune Diseases/immunology , Cell Cycle Proteins/immunology , GTP-Binding Proteins , Receptors, Cell Surface/immunology , Receptors, G-Protein-Coupled , Animals , Autoimmune Diseases/genetics , Autoimmunity/immunology , B-Lymphocytes/immunology , Cell Cycle Proteins/genetics , Cell Division , Female , Lymph Nodes/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/immunology , Receptors, Cell Surface/genetics , T-Lymphocytes/immunology , Time Factors
13.
Int Immunol ; 13(4): 485-93, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11282988

ABSTRACT

Formation of the pre-BCR complex is a critical check point during B cell development and induces the transition of pro-B to pre-B cells. CD79b (Igbeta) is a signaling component in the pre-BCR complex, since differentiation to the pre-B phenotype is induced by cross-linking the CD79b expressed on developmentally arrested pro-B cells from recombination-activating gene (RAG)-2-deficient mice. Bruton's tyrosine kinase (BTK) plays important roles in B cell development. However, its molecular mechanisms in early B cell development are not fully understood. To examine whether BTK functions in CD79b-mediated signaling for the pro-B/pre-B transition, we utilized RAG2/BTK double-knockout (DKO) mice. Pro-B cells from RAG2/BTK-DKO mice did not differentiate into pre-B cells following CD79b cross-linking, although tyrosine phosphorylation of cellular proteins including Erk1/2 and phospholipase C-gamma2 was induced in the same manner as RAG2-KO mice. BTK is phosphorylated after cross-linking of CD79b on RAG2-deficient pro-B cells. These findings suggest that BTK-dependent pathways downstream of CD79b are critical for the pro-B/pre-B transition and BTK-independent signaling pathways are also activated via the pre-BCR complex.


Subject(s)
Antigens, CD/immunology , B-Lymphocytes/immunology , Cell Differentiation , Protein-Tyrosine Kinases/physiology , Signal Transduction , Agammaglobulinaemia Tyrosine Kinase , Animals , B-Lymphocytes/cytology , B-Lymphocytes/enzymology , CD79 Antigens , DNA-Binding Proteins , Isoenzymes/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Phospholipase C gamma , Phosphorylation , Protein-Tyrosine Kinases/genetics , Receptors, Antigen, B-Cell/immunology , Type C Phospholipases/metabolism , Tyrosine/metabolism
14.
Cancer Res ; 61(8): 3256-61, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11309275

ABSTRACT

Prostate Stem Cell Antigen (PSCA) is a glycosylphosphatidylinositol-anchored cell surface protein that is expressed in normal human prostate and overexpressed in human prostate cancers. To test whether different pathways that generate prostate cancer would affect PSCA expression, a murine model system was developed. Monoclonal antibodies were generated against murine PSCA (mPSCA). mPSCA is expressed on approximately 20% of cells in normal prostate epithelium, and this number decreases with increasing age. In the transgenic adenocarcinoma of the mouse prostate (TRAMP) model of prostate cancer, tumors develop between 19 and 25 weeks of age. Murine PSCA was strongly expressed on approximately 60% of the cells of TRAMP tumors, at an age where the number of PSCA+ cells and the level of expression of PSCA is very low in the normal prostate. Phosphatase and tensin homologue deleted on chromosome 10 (PTEN) +/- mice develop a number of different cancers, including prostate cancer. The incidence of prostate cancer is low and occurs after a relatively long latency. Fluorescence-activated cell sorter analysis of prostatic tissue from 11-18-month-old PTEN +/- mice showed elevated numbers of PSCA+ cells in the prostate, and immunohistochemical analysis showed high mPSCA expression in the tumors of these mice. Together, these results show that two distinct mechanisms of carcinogenesis lead to expression of a common target antigen.


Subject(s)
Adenocarcinoma/immunology , Membrane Glycoproteins/biosynthesis , Neoplasm Proteins/biosynthesis , Prostatic Neoplasms/immunology , Tumor Suppressor Proteins , Adenocarcinoma/metabolism , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Antigens, Neoplasm , Cricetinae , Cricetulus , Female , GPI-Linked Proteins , Glutathione Transferase/biosynthesis , Glutathione Transferase/genetics , Humans , Immunohistochemistry , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Orchiectomy , PTEN Phosphohydrolase , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/physiology , Prostatic Neoplasms/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Testosterone/blood
15.
Proc Natl Acad Sci U S A ; 98(5): 2658-63, 2001 Feb 27.
Article in English | MEDLINE | ID: mdl-11226295

ABSTRACT

Prostate stem-cell antigen (PSCA) is a cell-surface antigen expressed in normal prostate and overexpressed in prostate cancer tissues. PSCA expression is detected in over 80% of patients with local disease, and elevated levels of PSCA are correlated with increased tumor stage, grade, and androgen independence, including high expression in bone metastases. We evaluated the therapeutic efficacy of anti-PSCA mAbs in human prostate cancer xenograft mouse models by using the androgen-dependent LAPC-9 xenograft and the androgen-independent recombinant cell line PC3-PSCA. Two different anti-PSCA mAbs, 1G8 (IgG1kappa) and 3C5 (IgG2akappa), inhibited formation of s.c. and orthotopic xenograft tumors in a dose-dependent manner. Furthermore, administration of anti-PSCA mAbs led to retardation of established orthotopic tumor growth and inhibition of metastasis to distant sites, resulting in a significant prolongation in the survival of tumor-bearing mice. These studies suggest PSCA as an attractive target for immunotherapy and demonstrate the therapeutic potential of anti-PSCA mAbs for the treatment of local and metastatic prostate cancer.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , Cell Division/immunology , Membrane Glycoproteins/immunology , Neoplasm Metastasis/immunology , Neoplasm Proteins/immunology , Prostatic Neoplasms/pathology , Animals , GPI-Linked Proteins , Humans , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Male , Mice , Mice, SCID , Neoplasm Transplantation , Survival Analysis , Tumor Cells, Cultured
16.
Stem Cells ; 18(6): 399-408, 2000.
Article in English | MEDLINE | ID: mdl-11072027

ABSTRACT

Chronic myeloid leukemia (CML) was the first human malignancy shown to be associated with a specific cytogenetic lesion, the Philadelphia chromosomal translocation. Forty years on, many biological and biochemical properties have been ascribed to its molecular product, the BCR-ABL tyrosine kinase fusion protein. However, it has been difficult to establish their precise contribution to the deregulation of normal survival, proliferative and differentiative control in chronic phase CML and the degree to which the involvement of stem cells extends beyond their role as the aetiological target. This review will focus on our current understanding of the pathogenesis of CML from the perspective of stem cell involvement, and how the biological and biochemical properties ascribed to BCR-ABL from studies of in vitro transformation and in vivo leukemogenesis systems relate to the abnormalities manifest in the human disease.


Subject(s)
Fusion Proteins, bcr-abl/biosynthesis , Hematopoietic Stem Cells/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Animals , Disease Models, Animal , Humans
17.
Proc Natl Acad Sci U S A ; 97(22): 12233-8, 2000 Oct 24.
Article in English | MEDLINE | ID: mdl-11027300

ABSTRACT

Cellular transformation by the BCR/ABL oncogene depends on the ABL-encoded tyrosine kinase activity. To block BCR/ABL function, we created a unique tyrosine phosphatase by fusing the catalytic domain of SHP1 (SHP1c) to the ABL binding domain (ABD) of RIN1, an established binding partner and substrate for c-ABL and BCR/ABL. This fusion construct (ABD/SHP1c) binds to BCR/ABL in cells and functions as an active phosphatase. ABD/SHP1c effectively suppressed BCR/ABL function as judged by reductions in transformation of fibroblast cells, growth factor independence of hematopoietic cell lines, and proliferation of primary bone marrow cells. In addition, the leukemogenic properties of BCR/ABL in a murine model system were blocked by coexpression of ABD/SHP1c. Both the "escort" function provided by ABD and the inhibitor function provided by the phosphatase of SHP1c were necessary for effective BCR/ABL interference. Expression of ABD/SHP1c also reversed the transformed phenotype of K562, a human leukemia-derived cell line. These results have direct implications for leukemia therapeutics and suggest an approach to block aberrant signal transduction in other pathologies through the use of appropriately designed escort/inhibitors.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Protein Tyrosine Phosphatases/metabolism , Recombinant Fusion Proteins/metabolism , Animals , Base Sequence , DNA Primers , Humans , Intracellular Signaling Peptides and Proteins , Leukemia/pathology , Mice , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Signal Transduction , Subcellular Fractions/metabolism , Tyrosine/metabolism
18.
Proc Natl Acad Sci U S A ; 97(22): 12109-14, 2000 Oct 24.
Article in English | MEDLINE | ID: mdl-11050239

ABSTRACT

G2A is an orphan G protein-coupled receptor (GPCR), expressed predominantly in T and B cells and homologous to a small group of GPCRs of unknown function expressed in lymphoid tissues. G2A is transcriptionally induced in response to diverse stimuli, and its ectopic expression suppresses transformation of B lymphoid precursors by BCR-ABL. G2A induces morphological transformation of NIH 3T3 fibroblasts. Microinjection of constructs encoding G2A into Swiss 3T3 fibroblasts induces actin reorganization into stress fibers that depends on RhoA, but not CDC42 or RAC. G2A elicits RhoA-dependent transcriptional activation of serum response factor. Direct evaluation of RhoA activity demonstrates elevated levels of RhoA-GTP in G2A-expressing cells. Microinjection of embryonic fibroblasts derived from various G alpha knockout mice establishes a requirement for G alpha 13 but not G alpha 12 or G alpha q/11 in G2A-induced actin rearrangement. In conclusion, G2A represents a family of GPCRs expressed in lymphocytes that may link diverse stimuli to cytoskeletal reorganization and transcriptional activation through a pathway involving G alpha 13 and RhoA.


Subject(s)
Actins/metabolism , Cell Cycle Proteins/metabolism , GTP-Binding Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , rhoA GTP-Binding Protein/metabolism , Animals , Cell Line , Cytoskeleton/metabolism , Humans , Mice , Transcriptional Activation
20.
Immunol Rev ; 175: 120-7, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10933597

ABSTRACT

Mutations in Bruton's tyrosine kinase (Btk) result in the B-cell immunodeficiencies X-linked agammaglobulinemia in humans and X-linked immunodeficiency in mice. These diseases are characterized by blocks in B-cell development at multiple stages and impaired function of residual mature B cells. This review focuses on a series of in vivo genetic studies that have begun to define the mechanism by which Btk regulates B-cell development and function. The functional interactions between Btk and other signaling molecules defined by this approach are more complex than initially appreciated from in vitro biochemical and cell culture studies.


Subject(s)
B-Lymphocytes/immunology , Protein-Tyrosine Kinases/physiology , Agammaglobulinaemia Tyrosine Kinase , Agammaglobulinemia/immunology , Animals , Cell Differentiation , Mice , Mice, Knockout , Mutation , Protein-Tyrosine Kinases/genetics , Signal Transduction , Transgenes , X Chromosome/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...