Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Cogn Emot ; 37(4): 748-762, 2023.
Article in English | MEDLINE | ID: mdl-37104122

ABSTRACT

Recognising and responding appropriately to emotions is critical to adaptive psychological functioning. Psychopathic traits (e.g. callous, manipulative, impulsive, antisocial) are related to differences in recognition and response when emotion is conveyed through facial expressions and language. Use of emotional music stimuli represents a promising approach to improve our understanding of the specific emotion processing difficulties underlying psychopathic traits because it decouples recognition of emotion from cues directly conveyed by other people (e.g. facial signals). In Experiment 1, participants listened to clips of emotional music and identified the emotional content (Sample 1, N = 196) or reported on their feelings elicited by the music (Sample 2, N = 197). Participants accurately recognised (t(195) = 32.78, p < .001, d = 4.69) and reported feelings consistent with (t(196) = 7.84, p < .001, d = 1.12) the emotion conveyed in the music. However, psychopathic traits were associated with reduced emotion recognition accuracy (F(1, 191) = 19.39, p < .001) and reduced likelihood of feeling the emotion (F(1, 193) = 35.45, p < .001), particularly for fearful music. In Experiment 2, we replicated findings for broad difficulties with emotion recognition (Sample 3, N = 179) and emotional resonance (Sample 4, N = 199) associated with psychopathic traits. Results offer new insight into emotion recognition and response difficulties that are associated with psychopathic traits.


Subject(s)
Music , Humans , Antisocial Personality Disorder/psychology , Emotions/physiology , Fear , Facial Expression
2.
Leukemia ; 31(4): 853-860, 2017 04.
Article in English | MEDLINE | ID: mdl-27773931

ABSTRACT

MYC is a major oncogenic driver of multiple myeloma (MM) and yet almost no therapeutic agents exist that target MYC in MM. Here we report that the let-7 biogenesis inhibitor LIN28B correlates with MYC expression in MM and is associated with adverse outcome. We also demonstrate that the LIN28B/let-7 axis modulates the expression of MYC, itself a let-7 target. Further, perturbation of the axis regulates the proliferation of MM cells in vivo in a xenograft tumor model. RNA-sequencing and gene set enrichment analyses of CRISPR-engineered cells further suggest that the LIN28/let-7 axis regulates MYC and cell cycle pathways in MM. We provide proof of principle for therapeutic regulation of MYC through let-7 with an LNA-GapmeR (locked nucleic acid-GapmeR) containing a let-7b mimic in vivo, demonstrating that high levels of let-7 expression repress tumor growth by regulating MYC expression. These findings reveal a novel mechanism of therapeutic targeting of MYC through the LIN28B/let-7 axis in MM that may impact other MYC-dependent cancers as well.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Multiple Myeloma/genetics , RNA Interference , RNA-Binding Proteins/genetics , Animals , Case-Control Studies , Cell Cycle/genetics , Cell Line, Tumor , Cluster Analysis , Disease Models, Animal , Gene Expression Profiling , Genes, myc , Heterografts , Humans , Mice , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Prognosis , RNA-Binding Proteins/metabolism
3.
Leukemia ; 29(6): 1320-30, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25655194

ABSTRACT

Mast cells (MCs) are critical components of the innate immune system and important for host defense, allergy, autoimmunity, tissue regeneration and tumor progression. Dysregulated MC development leads to systemic mastocytosis (SM), a clinically variable but often devastating family of hematologic disorders. Here we report that induced expression of Lin28, a heterochronic gene and pluripotency factor implicated in driving a fetal hematopoietic program, caused MC accumulation in adult mice in target organs such as the skin and peritoneal cavity. In vitro assays revealed a skewing of myeloid commitment in LIN28B-expressing hematopoietic progenitors, with increased levels of LIN28B in common myeloid and basophil-MC progenitors altering gene expression patterns to favor cell fate choices that enhanced MC specification. In addition, LIN28B-induced MCs appeared phenotypically and functionally immature, and in vitro assays suggested a slowing of MC terminal differentiation in the context of LIN28B upregulation. Finally, interrogation of human MC leukemia samples revealed upregulation of LIN28B in abnormal MCs from patients with SM. This work identifies Lin28 as a novel regulator of innate immune function and a new protein of interest in MC disease.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/physiology , Leukemia, Mast-Cell/pathology , Mast Cells/cytology , Mastocytosis, Systemic/pathology , Myeloid Cells/cytology , RNA-Binding Proteins/metabolism , Aged , Aged, 80 and over , Animals , Blotting, Western , Bone Marrow Transplantation , Cells, Cultured , Female , Flow Cytometry , Hematopoiesis/physiology , Humans , Leukemia, Mast-Cell/metabolism , Leukemia, Mast-Cell/therapy , Male , Mast Cells/metabolism , Mastocytosis, Systemic/metabolism , Mastocytosis, Systemic/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myeloid Cells/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
4.
Int J STD AIDS ; 25(7): 475-87, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24517928

ABSTRACT

Mycoplasma genitalium (M. genitalium)was first isolated from the urethral swabs of two symptomatic men with urethritis in 1980. Published prevalence rates vary greatly between populations studied. A number of urogenital conditions have been ascribed toM. genitalium, which is recognised to cause a sexually transmitted infection. The association of M. genitalium with non-specific urethritis is now well established, but the evidence supporting its role in both male and female infertility remains inconclusive. Laboratory methods are challenging and there is a lack of test standardisation. The recommended treatment of the infection is azithromycin as a single 1 gm dose. However, in recent years macrolide resistance has been observed. More studies are required to establish the clinical importance of M. genitaliumin urogenital conditions, particularly infertility, and to establish the role for screening and treatment in high-risk populations.


Subject(s)
Mycoplasma Infections/microbiology , Mycoplasma genitalium , Sexually Transmitted Diseases/microbiology , Urethritis/microbiology , Anti-Bacterial Agents/therapeutic use , DNA, Bacterial , Female , Humans , Macrolides/therapeutic use , Male , Mycoplasma Infections/drug therapy , Mycoplasma genitalium/drug effects , Mycoplasma genitalium/genetics , Mycoplasma genitalium/isolation & purification , Sexually Transmitted Diseases/drug therapy , Sexually Transmitted Diseases/transmission , Urethritis/drug therapy
5.
Leuk Suppl ; 3(Suppl 1): S3-4, 2014 Dec.
Article in English | MEDLINE | ID: mdl-27175271
6.
Nature ; 467(7313): 285-90, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20644535

ABSTRACT

Somatic cell nuclear transfer and transcription-factor-based reprogramming revert adult cells to an embryonic state, and yield pluripotent stem cells that can generate all tissues. Through different mechanisms and kinetics, these two reprogramming methods reset genomic methylation, an epigenetic modification of DNA that influences gene expression, leading us to hypothesize that the resulting pluripotent stem cells might have different properties. Here we observe that low-passage induced pluripotent stem cells (iPSCs) derived by factor-based reprogramming of adult murine tissues harbour residual DNA methylation signatures characteristic of their somatic tissue of origin, which favours their differentiation along lineages related to the donor cell, while restricting alternative cell fates. Such an 'epigenetic memory' of the donor tissue could be reset by differentiation and serial reprogramming, or by treatment of iPSCs with chromatin-modifying drugs. In contrast, the differentiation and methylation of nuclear-transfer-derived pluripotent stem cells were more similar to classical embryonic stem cells than were iPSCs. Our data indicate that nuclear transfer is more effective at establishing the ground state of pluripotency than factor-based reprogramming, which can leave an epigenetic memory of the tissue of origin that may influence efforts at directed differentiation for applications in disease modelling or treatment.


Subject(s)
Epigenesis, Genetic , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cellular Reprogramming/genetics , DNA Methylation/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genome/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Nuclear Transfer Techniques , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Article in English | MEDLINE | ID: mdl-19150965

ABSTRACT

With its hallmarks of unregulated cell proliferation and compromised differentiation, cancer represents a derangement of normal tissue homeostasis. A common set of pathways are activated in the transformed state, through either mutation or altered epigenetic regulation, and both heritable effects sustain the tumor. Classical views of cancer have invoked tissue dedifferentiation in the oncogenic process, whereas modern views embodied in the cancer stem cell hypothesis hold that cancer emerges from primitive tissue stem cells or specific progenitor populations that through mutations assume the self-renewal properties of stem cells. Recently, somatic tissues have been reprogrammed to a pluripotent state resembling embryonic stem (ES) cells by ectopic expression of a cocktail of transcription factors. The factors that drive reprogramming are oncogenes or have been linked to cellular transformation, suggesting that tumorigenesis and somatic cell reprogramming might indeed share common mechanisms of dedifferentiation.


Subject(s)
Cell Differentiation/genetics , Cell Differentiation/physiology , Neoplasms/genetics , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Epigenesis, Genetic , Female , Humans , Male , Neoplasms/physiopathology , Neoplastic Stem Cells/physiology , Pluripotent Stem Cells/pathology , Pluripotent Stem Cells/physiology
9.
Oncogene ; 27(6): 775-82, 2008 Jan 31.
Article in English | MEDLINE | ID: mdl-17684485

ABSTRACT

Resistance to molecularly targeted chemotherapy, and the development of novel agents that are active against resistant forms of target proteins create the need for a sensitive and quantitative assay to monitor drug-resistant mutations in patients to guide treatment and assess response. Here, we describe an application of the polymerase colony (polony) method to identify and quantify known point mutations in the BCR-ABL oncogene in patients with chronic myelogenous leukemia who evolve resistance to ABL kinase inhibitors. The assay can detect mutations with a sensitivity of 10(-4), quantify the burden of drug-resistant cells, and simultaneously monitor the dynamics of several coexisting mutations. As a proof of concept, we analysed blood samples from three patients undergoing therapy with ABL kinase inhibitors and found that the patients' response to therapy correlated with our molecular monitoring. We were also able to detect mutations emerging in patients long before clinical relapse. Therefore, the polony assay could be applied to a larger patient sample to assess the utility of early mutation detection in patient-specific treatment decisions. Finally, this methodology could be a valuable research tool to shed light on the natural behavior of mutations pre-existing kinase inhibitors therapy and either disappearing over time or slowly taking over.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Mutational Analysis/methods , Piperazines/pharmacology , Polymerase Chain Reaction/methods , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/genetics , Pyrimidines/pharmacology , Antineoplastic Agents/therapeutic use , Benzamides , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Mutation , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/therapeutic use
10.
Gene Ther ; 13(20): 1431-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17016468

ABSTRACT

With the isolation of human embryonic stem cells (hESCs) in 1998 came the realization of a long-sought aspiration for an unlimited source of human tissue. The difficulty of differentiating ESCs to pure, clinically exploitable cell populations to treat genetic and degenerative diseases is being solved in part with the help of genetically modified cell lines. With progress in genome editing and somatic cell nuclear transfer, it is theoretically possible to obtain genetically repaired isogenic cells. Moreover, the prospect of being able to select, isolate and expand a single cell to a vast population of cells could achieve a unique level of quality control, until now unattainable in the field of gene therapy. Most of the tools necessary to develop these strategies already exist in the mouse ESC system. We review here the advances accomplished in those fields and present some possible applications to hESC research.


Subject(s)
Cell Line , Embryonic Stem Cells , Genetic Diseases, Inborn/therapy , Genetic Therapy/trends , Animals , Gene Transfer Techniques , Genetic Therapy/methods , Humans , Immune Tolerance , Mice , Models, Animal , Pluripotent Stem Cells , Recombination, Genetic , Stem Cell Transplantation
11.
Cell Mol Life Sci ; 63(7-8): 760-6, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16568245

ABSTRACT

Embryonic stem cells provide an in vitro model for developmental biologists to study cell fate decisions during ontogenesis, while somatic stem cells allow physiologists to understand tissue homeostasis in the adult. The behavior of stem cells is dependent on an intimate relationship with a supportive niche. This brief review highlights some of the most important recent trends in stem cell biology, focusing in particular on the supportive microenvironments for both embryonic and adult stem cells. Known intrinsic and extrinsic molecular players from the best-characterized stem cell types are summarized, illuminating a number of shared environmental cues among tissues originating from all three embryonic germ layers.


Subject(s)
Stem Cells , Animals , Cell Differentiation , Cell Proliferation , Humans , Models, Biological , Signal Transduction , Stem Cells/physiology
12.
Leukemia ; 17(12): 2358-82, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14562120

ABSTRACT

Novel approaches have been designed to treat leukemia based on our understanding of the genetic and biochemical lesions present in different malignancies. This meeting report summarizes some of the recent advances in leukemia treatment. Based on the discoveries of cellular oncogenes, chromosomal translocations, monoclonal antibodies, multidrug resistance pumps, signal transduction pathways, genomics/proteonomic approaches to clinical diagnosis and mutations in biochemical pathways, clinicians and basic scientists have been able to identify the particular genetic mutations and signal transduction pathways involved as well as design more appropriate treatments for the leukemia patient. This meeting report discusses these exciting new therapies and the results obtained from ongoing clinical trials. Furthermore, rational approaches to treat complications of tumor lysis syndrome by administration of the recombinant urate oxidase protein, also known as rasburicase, which corrects the biochemical defect present in humans, were discussed. Clearly, over the past 25 years, molecular biology and biotechnology has provided the hematologist/oncologist novel bullets in their arsenal that will allow treatment by design in leukemia.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia/drug therapy , Leukemia/physiopathology , Medical Oncology/trends , Humans
13.
Circulation ; 104(22): 2641-4, 2001 Nov 27.
Article in English | MEDLINE | ID: mdl-11723011

ABSTRACT

BACKGROUND: Recent advances in high-throughput genomics technology have expanded our ability to catalogue allelic variants in large sets of candidate genes related to premature coronary artery disease. METHODS AND RESULTS: A total of 398 families were identified in 15 participating medical centers; they fulfilled the criteria of myocardial infarction, revascularization, or a significant coronary artery lesion diagnosed before 45 years in men or 50 years in women. A total of 62 vascular biology genes and 72 single-nucleotide polymorphisms were assessed. Previously undescribed variants in 3 related members of the thrombospondin protein family were prominent among a small set of single-nucleotide polymorphisms that showed a statistical association with premature coronary artery disease. A missense variant of thrombospondin 4 (A387P) showed the strongest association, with an adjusted odds ratio for myocardial infarction of 1.89 (P=0.002 adjusted for covariates) for individuals carrying the P allele. A variant in the 3' untranslated region of thrombospondin-2 (change of thymidine to guanine) seemed to have a protective effect against myocardial in individuals homozygous for the variant (adjusted odds ratio of 0.31; P=0.0018). A missense variant in thrombospondin-1 (N700S) was associated with an adjusted odds ratio for coronary artery disease of 11.90 (P=0.041) in homozygous individuals, who also had the lowest level of thrombospondin-1 by plasma assay (P=0.0019). CONCLUSIONS: This large-scale genetic study has identified the potential of multiple novel variants in the thrombospondin gene family to be associated with familial premature myocardial infarction. Notwithstanding multiple caveats, thrombospondins specifically and high-throughput genomic technology in general deserve further study in familial ischemic heart disease.


Subject(s)
Coronary Artery Disease/genetics , Myocardial Infarction/diagnosis , Myocardial Infarction/genetics , Polymorphism, Single Nucleotide/genetics , Thrombospondins/genetics , Adult , Age of Onset , Alleles , Case-Control Studies , Coronary Angiography , Coronary Artery Disease/epidemiology , Coronary Stenosis/diagnosis , Coronary Stenosis/genetics , Demography , Female , Genetic Predisposition to Disease , Genetic Testing , Genotype , Homozygote , Humans , Male , Methylenetetrahydrofolate Reductase (NADPH2) , Middle Aged , Myocardial Infarction/epidemiology , Odds Ratio , Oxidoreductases Acting on CH-NH Group Donors/genetics , Predictive Value of Tests , Thrombospondin 1/genetics , United States
14.
Development ; 128(22): 4597-604, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11714684

ABSTRACT

Embryonic stem (ES) cells differentiate into multiple hematopoietic lineages during embryoid body formation in vitro, but to date, an ES-derived hematopoietic stem cell has not been identified and subjected to clonal analysis in a manner comparable with hematopoietic stem cells from adult bone marrow. As the chronic myeloid leukemia-associated BCR/ABL oncogene endows the adult hematopoietic stem cell with clonal dominance without inhibiting pluripotent lymphoid and myeloid differentiation, we have used BCR/ABL as a tool to enable engraftment and clonal analysis. We show that embryoid body-derived hematopoietic progenitors expressing BCR/ABL maintain a primitive hematopoietic blast stage of differentiation and generate only primitive erythroid cell types in vitro. These cells can be cloned, and when injected into irradiated adult mice, they differentiate into multiple myeloid cell types as well as T and B lymphocytes. While the injected cells express embryonic (beta-H1) globin, donor-derived erythroid cells in the recipient express only adult (beta-major) globin, suggesting that these cells undergo globin gene switching and developmental maturation in vivo. These data demonstrate that an embryonic hematopoietic stem cell arises in vitro during ES cell differentiation that constitutes a common progenitor for embryonic erythroid and definitive lymphoid-myeloid hematopoiesis.


Subject(s)
Embryo, Mammalian/cytology , Erythropoiesis , Fusion Proteins, bcr-abl/genetics , Hematopoietic Stem Cells/cytology , Leukopoiesis , Animals , Bone Marrow Cells , Cell Differentiation , Clone Cells , Hematopoietic Stem Cell Transplantation , Mice , Models, Biological , Radiation Chimera , Spleen/cytology , Transformation, Genetic
15.
Oncogene ; 20(41): 5826-35, 2001 Sep 13.
Article in English | MEDLINE | ID: mdl-11593388

ABSTRACT

The Akt, Ras and STAT5 signaling pathways have each been linked to transformation of hematopoietic cells by BCR/ABL. However the relative contributions of these signaling pathways to BCR/ABL mediated cytokine-independent survival, proliferation and resistance to DNA damage-induced apoptosis have not been systematically defined. Here we report that activation of either Akt, Ras or STAT5 confers cytokine-independent survival to IL-3 dependent BaF3 cells. Ras or STAT5, but not Akt, also drives cytokine-independent proliferation and imparts sustained resistance to DNA damage-induced apoptosis. We also show that dominant negative (DN) inhibition of STAT5, but not Ras or Akt, significantly reduces resistance to DNA damage-induced apoptosis in BCR/ABL transformed BaF3 cells. Whereas inhibition of STAT5 or Ras alone does not compromise cytokine-independent proliferation of BaF3-BCR/ABL cells, simultaneous blockade of both STAT5 and Ras reduces proliferation and maximally sensitizes BaF3-BCR/ABL cells to DNA damage induced by gamma-irradiation, suggesting a cooperative role for these two signaling pathways in BCR/ABL transformation. The anti-apoptotic properties of BCR/ABL can be partly explained by an increase in the expression of Pim-1 and Bcl-XL, as ectopic expression of these STAT5 target genes imparts both cytokine-independent survival and partial gamma-radiation resistance. These data illustrate both cooperative and redundant effects of STAT5 and Ras signaling in BCR/ABL transformed cells, with STAT5 playing a dominant role in resistance to DNA damage-induced apoptosis.


Subject(s)
DNA-Binding Proteins/physiology , Fusion Proteins, bcr-abl/physiology , Hematopoietic Stem Cells/physiology , Milk Proteins , Protein Serine-Threonine Kinases/physiology , Trans-Activators/physiology , ras Proteins/physiology , Animals , Apoptosis/physiology , Cell Division , Cell Survival/drug effects , DNA Damage , DNA-Binding Proteins/genetics , Fusion Proteins, bcr-abl/genetics , Genetic Vectors/genetics , Hematopoietic Stem Cells/drug effects , Interleukin-3/pharmacology , Philadelphia Chromosome , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-pim-1 , Retroviridae , STAT5 Transcription Factor , Signal Transduction , Trans-Activators/genetics , Transduction, Genetic , bcl-X Protein , ras Proteins/genetics
16.
Trends Cardiovasc Med ; 11(2): 60-6, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11530294

ABSTRACT

Several environmental risk factors of cardiovascular disease are well established, but genetic risk alleles contributing to the disease in the general population are hotly debated. New strategies focusing on polymorphism discovery in candidate disease genes followed by tests of association to genes across the genome offer a pioneering approach to identifying risk alleles. Several hundred candidate genes for cardiovascular disease have been screened for common polymorphisms and these variants may provide susceptibility alleles which largely contribute to risk of cardiovascular disease in the general population. However, the impact of common susceptibility alleles for disease management will depend on many years of future investigation.


Subject(s)
Heart/physiology , Myocardium/metabolism , Polymorphism, Single Nucleotide/genetics , Cardiovascular Diseases/genetics , Family Health , Humans , Polymorphism, Genetic/genetics
17.
Oncogene ; 20(21): 2636-46, 2001 May 10.
Article in English | MEDLINE | ID: mdl-11420675

ABSTRACT

During differentiation in vitro, Embryonic Stem (ES) cells generate both primitive erythroid and definitive myeloid lineages in a process that mimics hematopoiesis in the mammalian yolk sac. To investigate leukemic transformation of these embryonic hematopoietic progenitors, we infected differentiating cultures of ES cells with the Chronic Myeloid Leukemia-specific BCR/ABL oncoprotein. Following a period of liquid culture, we isolated two transformed subclones, EB57 and EB67, that retained characteristics of embryonic hematopoietic progenitors and induced a fatal leukemia in mice characterized by massive splenomegaly and granulocytosis. Histopathology of the spleen revealed an abundance of undifferentiated blast-like cells. Investigation of the clonal origins of the granulocytes in the peripheral blood demonstrated that the injected donor cells contributed modestly to the granulocyte population while the majority were host-derived. EB57 secretes IL-3 and unidentified cytokines that can stimulate autocrine and paracrine cell proliferation, presumably accounting for the reactive granulocytosis in diseased mice. These BCR/ABL transformed hematopoietic derivatives of ES cells recapitulate the relationship of BCR/ABL expression to IL-3 production that has been described for primitive hematopoietic progenitors from human CML patients, and illustrates the potential for autocrine and paracrine effects of BCR/ABL-infected cells in murine models.


Subject(s)
Cell Transformation, Viral/genetics , Fusion Proteins, bcr-abl/physiology , Genes, abl , Hematopoietic Stem Cells/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Animals , Antigens, Surface/biosynthesis , Cell Differentiation/physiology , Cell Line, Transformed , Erythroid Precursor Cells/cytology , Fusion Proteins, bcr-abl/biosynthesis , Fusion Proteins, bcr-abl/genetics , Granulocytes/pathology , Hematopoietic Stem Cells/physiology , Interleukin-3/biosynthesis , Interleukin-3/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Mice, Inbred BALB C , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction
18.
Blood ; 97(11): 3491-7, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11369642

ABSTRACT

Mice deficient in the interferon consensus sequence binding protein (ICSBP) develop a disease resembling chronic myeloid leukemia (CML), which in humans is caused by the BCR/ABL oncoprotein. Interferon-alpha (IFN-alpha) induces ICSBP expression and is an effective therapy for CML. This study examined whether enforced expression of ICSBP might antagonize BCR/ABL-induced leukemia; results demonstrated that ICSBP-modified cells generated a protective CD8(+) cytotoxic T-cell response against BCR/ABL-transformed BaF3 cells in a murine leukemia model. ICSBP expression represents a novel means of stimulating a host immune response to BCR/ABL(+) leukemia cells and a potential strategy for immunotherapy of CML. (Blood. 2001;97:3491-3497)


Subject(s)
Fusion Proteins, bcr-abl/physiology , Gene Expression , Leukemia/immunology , Repressor Proteins/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Transformation, Neoplastic , Female , Fusion Proteins, bcr-abl/analysis , Fusion Proteins, bcr-abl/genetics , Immunity , Immunization , Interferon Regulatory Factors , Interferons/physiology , Leukemia/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Repressor Proteins/physiology , Reverse Transcriptase Polymerase Chain Reaction , Spleen/immunology , Transfection
19.
Blood ; 97(5): 1404-12, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11222387

ABSTRACT

BCR/ABL, the oncoprotein responsible for chronic myeloid leukemia (CML), transforms hematopoietic cells through both Ras-dependent and -independent mechanisms. Farnesyl protein transferase inhibitors (FTIs) were designed to block mutant Ras signaling, but they also inhibit the growth of transformed cells with wild-type Ras, implying that other farnesylated targets contribute to FTI action. In the current study, the clinical candidate FTI SCH66336 was characterized for its ability to inhibit BCR/ABL transformation. When tested against BCR/ABL-BaF3 cells, a murine cell line that is leukemogenic in mice, SCH66336 potently inhibited soft agar colony formation, slowed proliferation, and sensitized cells to apoptotic stimuli. Quantification of activated guanosine triphosphate (GTP)-bound Ras protein and electrophoretic mobility shift assays for AP-1 DNA binding showed that Ras effector pathways are inhibited by SCH66336. However, SCH66336 was more inhibitory than dominant-negative Ras in assays of soft agar colony formation and cell proliferation, suggesting activity against targets other than Ras. Cell cycle analysis of BCR/ABL-BaF3 cells treated with SCH66336 revealed G2/M blockade, consistent with recent reports that centromeric proteins that regulate the G2/M checkpoint are critical farnesylated targets of FTI action. Mice injected intravenously with BCR/ABL-BaF3 cells developed acute leukemia and died within 4 weeks with massive splenomegaly, elevated white blood cell counts, and anemia. In contrast, nearly all mice treated with SCH66336 survived and have remained disease-free for more than a year. Furthermore, SCH66336 selectively inhibited the hematopoietic colony formation of primary human CML cells. As an oral, nontoxic compound with a mechanism of action distinct from that of ABL tyrosine kinase inhibition, FTI SCH66336 shows promise for the treatment of BCR/ABL-induced leukemia.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperidines/pharmacology , Pyridines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Cell Cycle/drug effects , Cell Division/drug effects , Farnesyltranstransferase , Genes, abl/physiology , Hematopoietic Stem Cells/drug effects , Humans , Leukemia, Experimental/drug therapy , Leukemia, Experimental/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Spleen/drug effects , Spleen/pathology , Survival Rate , Transformation, Genetic , Tumor Cells, Cultured
20.
Blood ; 97(5): 1399-403, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11222386

ABSTRACT

The Philadelphia (Ph) chromosome is found in approximately 3% of pediatric patients with acute lymphoblastic leukemia (ALL) and the percentage markedly increases in adult patients. The prognosis for this class of patients is poor, and no standard chemotherapy combination so far has demonstrated long-term efficacy. The Ph-translocation joins the BCR and ABL genes and leads to expression of a chimeric Bcr/Abl protein with enhanced tyrosine kinase activity. This increase in activity leads to malignant transformation by interference with basic cellular functions such as the control of proliferation, adherence to stroma and extracellular matrix, and apoptosis. One important pathway activated by Bcr/Abl is the Ras pathway. Ras proteins have to undergo a series of posttranslational modifications to become biologically active. The first modification is the farnesylation of the C-terminus catalyzed by farnesyl transferase. We studied the effect of the farnesyl transferase inhibitor SCH66336 in an in vivo murine model of Bcr/Abl-positive acute lymphoblastic leukemia. In the early leukemic phase, mice were randomly assigned to a treatment, a vehicle, and a nontreatment group. The treatment was well tolerated without any detectable side effects. All animals of the control groups died of leukemia/lymphoma within 103 days (range, 18-103 days). In contrast, 80% of the drug-receiving group survived without any signs of leukemia or lymphoma until termination of treatment, after a median treatment period of 200 days (range, 179-232 days). We conclude that farnesyl transferase inhibitor SCH66336 is able to revert early signs of leukemia and significantly prolongs survival in a murine ALL model.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Genes, abl/genetics , Piperidines/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Pyridines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bone Marrow , Farnesyltranstransferase , Genes, abl/drug effects , Mice , Mice, Transgenic , Piperidines/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Pyridines/therapeutic use , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...