Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Bone Marrow Transplant ; 59(3): 366-372, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38177222

ABSTRACT

ZUMA-1 safety management cohort 6 investigated the impact of prophylactic corticosteroids and earlier corticosteroids and/or tocilizumab on the incidence and severity of cytokine release syndrome (CRS) and neurologic events (NEs) following axicabtagene ciloleucel (axi-cel) in patients with relapsed/refractory large B-cell lymphoma (R/R LBCL). Prior analyses of cohort 6 with limited follow-up demonstrated no Grade ≥3 CRS, a low rate of NEs, and high response rates, without negatively impacting axi-cel pharmacokinetics. Herein, long-term outcomes of cohort 6 (N = 40) are reported (median follow-up, 26.9 months). Since the 1-year analysis (Oluwole, et al. Blood. 2022;138[suppl 1]:2832), no new CRS was reported. Two new NEs occurred in two patients (Grade 2 dementia unrelated to axi-cel; Grade 5 axi-cel-related leukoencephalopathy). Six new infections and eight deaths (five progressive disease; one leukoencephalopathy; two COVID-19) occurred. Objective and complete response rates remained at 95% and 80%, respectively. Median duration of response and progression-free survival were reached at 25.9 and 26.8 months, respectively. Median overall survival has not yet been reached. Eighteen patients (45%) remained in ongoing response at data cutoff. With ≥2 years of follow-up, prophylactic corticosteroids and earlier corticosteroids and/or tocilizumab continued to demonstrate CRS improvement without compromising efficacy outcomes, which remained high and durable.


Subject(s)
Biological Products , Leukoencephalopathies , Lymphoma, Large B-Cell, Diffuse , Humans , Adrenal Cortex Hormones/therapeutic use , Biological Products/therapeutic use , Cytokine Release Syndrome , Immunotherapy, Adoptive , Antigens, CD19
2.
Blood Cancer Discov ; 5(1): 21-33, 2024 01 08.
Article in English | MEDLINE | ID: mdl-37983485

ABSTRACT

Treatment resistance and toxicities remain a risk following chimeric antigen receptor (CAR) T-cell therapy. Herein, we report pharmacokinetics, pharmacodynamics, and product and apheresis attributes associated with outcomes among patients with relapsed/refractory large B-cell lymphoma (LBCL) treated with axicabtagene ciloleucel (axi-cel) in ZUMA-7. Axi-cel peak expansion associated with clinical response and toxicity, but not response durability. In apheresis material and final product, a naive T-cell phenotype (CCR7+CD45RA+) expressing CD27 and CD28 associated with improved response durability, event-free survival, progression-free survival, and a lower number of prior therapies. This phenotype was not associated with high-grade cytokine release syndrome (CRS) or neurologic events. Higher baseline and postinfusion levels of serum inflammatory markers associated with differentiated/effector products, reduced efficacy, and increased CRS and neurologic events, thus suggesting targets for intervention. These data support better outcomes with earlier CAR T-cell intervention and may improve patient care by informing on predictive biomarkers and development of next-generation products. SIGNIFICANCE: In ZUMA-7, the largest randomized CAR T-cell trial in LBCL, a naive T-cell product phenotype (CCR7+CD45RA+) expressing CD27 and CD28 associated with improved efficacy, decreased toxicity, and a lower number of prior therapies, supporting earlier intervention with CAR T-cell therapy. In addition, targets for improvement of therapeutic index are proposed. This article is featured in Selected Articles from This Issue, p. 4.


Subject(s)
Immunotherapy, Adoptive , Lymphoma, Large B-Cell, Diffuse , Humans , Immunotherapy, Adoptive/adverse effects , CD28 Antigens , Receptors, CCR7 , Lymphoma, Large B-Cell, Diffuse/therapy , Research Personnel , Cytokine Release Syndrome , Leukocyte Common Antigens
3.
Blood ; 141(19): 2307-2315, 2023 05 11.
Article in English | MEDLINE | ID: mdl-36821768

ABSTRACT

In phase 2 of ZUMA-1, a single-arm, multicenter, registrational trial, axicabtagene ciloleucel (axi-cel) autologous anti-CD19 chimeric antigen receptor (CAR) T-cell therapy demonstrated durable responses at 2 years in patients with refractory large B-cell lymphoma (LBCL). Here, we assessed outcomes in ZUMA-1 after 5 years of follow-up. Eligible adults received lymphodepleting chemotherapy followed by axi-cel (2 × 106 cells per kg). Investigator-assessed response, survival, safety, and pharmacokinetics were assessed in patients who had received treatment. The objective response rate in these 101 patients was 83% (58% complete response rate); with a median follow-up of 63.1 months, responses were ongoing in 31% of patients at data cutoff. Median overall survival (OS) was 25.8 months, and the estimated 5-year OS rate was 42.6%. Disease-specific survival (excluding deaths unrelated to disease progression) estimated at 5 years was 51.0%. No new serious adverse events or deaths related to axi-cel were observed after additional follow-up. Peripheral blood B cells were detectable in all evaluable patients at 3 years with polyclonal B-cell recovery in 91% of patients. Ongoing responses at 60 months were associated with early CAR T-cell expansion. In conclusion, this 5-year follow-up analysis of ZUMA-1 demonstrates sustained overall and disease-specific survival, with no new safety signals in patients with refractory LBCL. Protracted B-cell aplasia was not required for durable responses. These findings support the curative potential of axi-cel in a subset of patients with aggressive B-cell lymphomas. This trial was registered at ClinicalTrials.gov, as #NCT02348216.


Subject(s)
Biological Products , Lymphoma, Large B-Cell, Diffuse , Receptors, Chimeric Antigen , Adult , Humans , Follow-Up Studies , Immunotherapy, Adoptive/adverse effects , Lymphoma, Large B-Cell, Diffuse/pathology , Antigens, CD19/therapeutic use
4.
J Clin Oncol ; 41(3): 555-567, 2023 01 20.
Article in English | MEDLINE | ID: mdl-35658525

ABSTRACT

PURPOSE: Brexucabtagene autoleucel (KTE-X19) autologous anti-CD19 chimeric antigen receptor (CAR) T-cell therapy is approved for the treatment of relapsed/refractory mantle cell lymphoma (MCL). Outcomes after a 3-year follow-up in the pivotal ZUMA-2 study of KTE-X19 in relapsed/refractory MCL are reported, including for subgroups by prior therapy (bendamustine and type of Bruton tyrosine kinase inhibitor [BTKi]) or high-risk characteristics. METHODS: Patients with relapsed/refractory MCL (one to five prior therapies, including prior BTKi exposure) received a single infusion of KTE-X19 (2 × 106 CAR T cells/kg). RESULTS: After a median follow-up of 35.6 months, the objective response rate among all 68 treated patients was 91% (95% CI, 81.8 to 96.7) with 68% complete responses (95% CI, 55.2 to 78.5); medians for duration of response, progression-free survival, and overall survival were 28.2 months (95% CI, 13.5 to 47.1), 25.8 months (95% CI, 9.6 to 47.6), and 46.6 months (95% CI, 24.9 to not estimable), respectively. Post hoc analyses showed that objective response rates and ongoing response rates were consistent among prespecified subgroups by prior BTKi exposure or high-risk characteristics. In an exploratory analysis, patients with prior bendamustine benefited from KTE-X19, but showed a trend toward attenuated T-cell functionality, with more impact of bendamustine given within 6 versus 12 months of leukapheresis. Late-onset toxicities were infrequent; only 3% of treatment-emergent adverse events of interest in ZUMA-2 occurred during this longer follow-up period. Translational assessments revealed associations with long-term benefits of KTE-X19 including high-peak CAR T-cell expansion in responders and the predictive value of minimal residual disease for relapse. CONCLUSION: These data, representing the longest follow-up of CAR T-cell therapy in patients with MCL to date, suggest that KTE-X19 induced durable long-term responses with manageable safety in patients with relapsed/refractory MCL and may also benefit those with high-risk characteristics.


Subject(s)
Lymphoma, Mantle-Cell , Receptors, Chimeric Antigen , Adult , Humans , Receptors, Chimeric Antigen/therapeutic use , Lymphoma, Mantle-Cell/drug therapy , Immunotherapy, Adoptive/adverse effects , Follow-Up Studies , Bendamustine Hydrochloride/therapeutic use , Neoplasm Recurrence, Local/etiology
5.
Mol Cancer Res ; 13(3): 502-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25368431

ABSTRACT

UNLABELLED: High-grade serous ovarian cancers (HGSOC) are characterized by widespread recurrent regions of copy-number gain and loss. Here, we interrogated 50 genes that are recurrently amplified in HGSOC and essential for cancer proliferation and survival in ovarian cancer cell lines. FRS2 is one of the 50 genes located on chromosomal region 12q15 that is focally amplified in 12.5% of HGSOC. We found that FRS2-amplified cancer cell lines are dependent on FRS2 expression, and that FRS2 overexpression in immortalized human cell lines conferred the ability to grow in an anchorage-independent manner and as tumors in immunodeficient mice. FRS2, an adaptor protein in the FGFR pathway, induces downstream activation of the Ras-MAPK pathway. These observations identify FRS2 as an oncogene in a subset of HGSOC that harbor FRS2 amplifications. IMPLICATIONS: These studies identify FRS2 as an amplified oncogene in a subset of HGSOC. FRS2 expression is essential to ovarian cancer cells that harbor 12q15 amplification.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cystadenocarcinoma, Serous/pathology , Gene Amplification , Membrane Proteins/genetics , Membrane Proteins/metabolism , Ovarian Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , Chromosomes, Human, Pair 12/genetics , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Female , Humans , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism
6.
Cancer Discov ; 4(4): 452-65, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24444711

ABSTRACT

Although the roles of mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) signaling in KRAS-driven tumorigenesis are well established, KRAS activates additional pathways required for tumor maintenance, the inhibition of which are likely to be necessary for effective KRAS-directed therapy. Here, we show that the IκB kinase (IKK)-related kinases Tank-binding kinase-1 (TBK1) and IKKε promote KRAS-driven tumorigenesis by regulating autocrine CCL5 and interleukin (IL)-6 and identify CYT387 as a potent JAK/TBK1/IKKε inhibitor. CYT387 treatment ablates RAS-associated cytokine signaling and impairs Kras-driven murine lung cancer growth. Combined CYT387 treatment and MAPK pathway inhibition induces regression of aggressive murine lung adenocarcinomas driven by Kras mutation and p53 loss. These observations reveal that TBK1/IKKε promote tumor survival by activating CCL5 and IL-6 and identify concurrent inhibition of TBK1/IKKε, Janus-activated kinase (JAK), and MEK signaling as an effective approach to inhibit the actions of oncogenic KRAS.


Subject(s)
Autocrine Communication , Benzamides/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Pyrimidines/pharmacology , Signal Transduction/drug effects , ras Proteins/genetics , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Chemokine CCL5/metabolism , Human Umbilical Vein Endothelial Cells , Humans , I-kappa B Proteins/metabolism , Interleukin-6/metabolism , Mice , Neoplasms, Experimental , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism
7.
Cancer Discov ; 3(9): 1044-57, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23764425

ABSTRACT

UNLABELLED: 3q26 is frequently amplified in several cancer types with a common amplified region containing 20 genes. To identify cancer driver genes in this region, we interrogated the function of each of these genes by loss- and gain-of-function genetic screens. Specifically, we found that TLOC1 (SEC62) was selectively required for the proliferation of cell lines with 3q26 amplification. Increased TLOC1 expression induced anchorage-independent growth, and a second 3q26 gene, SKIL (SNON), facilitated cell invasion in immortalized human mammary epithelial cells. Expression of both TLOC1 and SKIL induced subcutaneous tumor growth. Proteomic studies showed that TLOC1 binds to DDX3X, which is essential for TLOC1-induced transformation and affected protein translation. SKIL induced invasion through upregulation of SLUG (SNAI2) expression. Together, these studies identify TLOC1 and SKIL as driver genes at 3q26 and more broadly suggest that cooperating genes may be coamplified in other regions with somatic copy number gain. SIGNIFICANCE: These studies identify TLOC1 and SKIL as driver genes in 3q26. These observations provide evidence that regions of somatic copy number gain may harbor cooperating genes of different but complementary functions.


Subject(s)
Chromosomes, Human, Pair 3/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Transport Proteins/genetics , Neoplasm Invasiveness/genetics , Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Breast Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation , DEAD-box RNA Helicases/metabolism , DNA Copy Number Variations/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Amplification/genetics , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/genetics , Mammary Glands, Human/cytology , Membrane Transport Proteins/metabolism , Ovarian Neoplasms/genetics , Protein Binding , Proto-Oncogene Proteins/metabolism , RNA Interference , RNA, Small Interfering , Snail Family Transcription Factors , Transcription Factors/biosynthesis
8.
Cell Rep ; 3(3): 724-33, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23453969

ABSTRACT

IκB kinase ε (IKKε, IKBKE) is a key regulator of innate immunity and a breast cancer oncogene, amplified in ~30% of breast cancers, that promotes malignant transformation through NF-κB activation. Here, we show that IKKε is modified and regulated by K63-linked polyubiquitination at lysine 30 and lysine 401. Tumor necrosis factor alpha and interleukin-1ß stimulation induces IKKε K63-linked polyubiquitination over baseline levels in both macrophages and breast cancer cell lines, and this modification is essential for IKKε kinase activity, IKKε-mediated NF-κB activation, and IKKε-induced malignant transformation. Disruption of K63-linked ubiquitination of IKKε does not affect its overall structure but impairs the recruitment of canonical NF-κB proteins. A cIAP1/cIAP2/TRAF2 E3 ligase complex binds to and ubiquitinates IKKε. Altogether, these observations demonstrate that K63-linked polyubiquitination regulates IKKε activity in both inflammatory and oncogenic contexts and suggests an alternative approach to targeting this breast cancer oncogene.


Subject(s)
Cell Transformation, Neoplastic/metabolism , I-kappa B Kinase/metabolism , Inhibitor of Apoptosis Proteins/metabolism , TNF Receptor-Associated Factor 2/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Amino Acid Sequence , Animals , Cell Line, Tumor , Interleukin-1beta/metabolism , Lysine/metabolism , Mice , Molecular Sequence Data , NF-kappa B/metabolism , Neoplasms, Experimental/metabolism , Tumor Necrosis Factor-alpha/metabolism
9.
Mol Cell Biol ; 32(23): 4756-68, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23007157

ABSTRACT

NF-κB transcription factors are central regulators of inflammation and when dysregulated contribute to malignant transformation. IκB kinase ε (IKKε; IKKi, encoded by IKBKE) is a breast oncogene that is amplified in 30% of breast cancers and drives transformation in an NF-κB-dependent manner. Here we demonstrate that IKKε interacts with and phosphorylates tumor necrosis factor receptor-associated factor 2 (TRAF2) at Ser11 in vitro and in vivo. This activity promotes Lys63-linked TRAF2 ubiquitination and NF-κB activation and is essential for IKKε transformation. Breast cancer cells that depend on IKKε expression for survival are also dependent on TRAF2. This work defines TRAF2 phosphorylation to be one key effector of IKKε-induced mammary epithelial cell transformation.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , I-kappa B Kinase/metabolism , TNF Receptor-Associated Factor 2/metabolism , Amino Acid Sequence , Animals , Breast/cytology , Breast/immunology , Breast/metabolism , Breast/pathology , Breast Neoplasms/immunology , Cell Line, Tumor , Cell Transformation, Neoplastic/immunology , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Deubiquitinating Enzyme CYLD , Down-Regulation , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , HEK293 Cells , Humans , I-kappa B Kinase/immunology , Mice , NF-kappa B/immunology , NIH 3T3 Cells , Nuclear Pore Complex Proteins/metabolism , Phosphorylation , RNA-Binding Proteins/metabolism , TNF Receptor-Associated Factor 2/chemistry , TNF Receptor-Associated Factor 2/immunology , Ubiquitination
10.
Mutagenesis ; 27(1): 93-101, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21852270

ABSTRACT

Cell transformation assays (CTAs) have long been proposed as in vitro methods for the identification of potential chemical carcinogens. Despite showing good correlation with rodent bioassay data, concerns over the subjective nature of using morphological criteria for identifying transformed cells and a lack of understanding of the mechanistic basis of the assays has limited their acceptance for regulatory purposes. However, recent drivers to find alternative carcinogenicity assessment methodologies, such as the Seventh Amendment to the EU Cosmetics Directive, have fuelled renewed interest in CTAs. Research is currently ongoing to improve the objectivity of the assays, reveal the underlying molecular changes leading to transformation and explore the use of novel cell types. The UK NC3Rs held an international workshop in November 2010 to review the current state of the art in this field and provide directions for future research. This paper outlines the key points highlighted at this meeting.


Subject(s)
Carcinogenicity Tests/methods , Carcinogens/toxicity , Animals , Biomarkers/analysis , Cell Line , Cell Transformation, Neoplastic , Congresses as Topic , Cosmetics/toxicity , Humans , Validation Studies as Topic
11.
Mol Cell ; 34(4): 461-72, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19481526

ABSTRACT

The noncanonical IKK family member IKKepsilon is essential for regulating antiviral signaling pathways and is a recently discovered breast cancer oncoprotein. Although several IKKepsilon targets have been described, direct IKKepsilon substrates necessary for regulating cell transformation have not been identified. Here, we performed a screen for putative IKKepsilon substrates using an unbiased proteomic and bioinformatic approach. Using a positional scanning peptide library assay, we determined the optimal phosphorylation motif for IKKepsilon and used bioinformatic approaches to predict IKKepsilon substrates. Of these potential substrates, serine 418 of the tumor suppressor CYLD was identified as a likely site of IKKepsilon phosphorylation. We confirmed that CYLD is directly phosphorylated by IKKepsilon and that IKKepsilon phosphorylates serine 418 in vivo. Phosphorylation of CYLD at serine 418 decreases its deubiquitinase activity and is necessary for IKKepsilon-driven transformation. Together, these observations define IKKepsilon and CYLD as an oncogene-tumor suppressor network that participates in tumorigenesis.


Subject(s)
Breast Neoplasms , Cell Transformation, Neoplastic , I-kappa B Kinase/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Deubiquitinating Enzyme CYLD , Female , Genes, Reporter , Humans , I-kappa B Kinase/genetics , Molecular Sequence Data , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation , Sequence Alignment , Serine/metabolism , Substrate Specificity , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , Tumor Suppressor Proteins/genetics
12.
Blood ; 113(11): 2478-87, 2009 Mar 12.
Article in English | MEDLINE | ID: mdl-19147787

ABSTRACT

B-cell lymphoma is the most common immune system malignancy. TCL1 transgenic mice (TCL1-tg), in which TCL1 is ectopically expressed in mature lymphocytes, develop multiple B- and T-cell leukemia and lymphoma subtypes, supporting an oncogenic role for TCL1 that probably involves AKT and MAPK-ERK signaling pathway augmentation. Additional, largely unknown genetic and epigenetic alterations cooperate with TCL1 during lymphoma progression. We examined DNA methylation patterns in TCL1-tg B-cell tumors to discover tumor-associated epigenetic changes, and identified hypermethylation of sprouty2 (Spry2). Sprouty proteins are context-dependent negative or positive regulators of MAPK-ERK pathway signaling, but their role(s) in B-cell physiology or pathology are unknown. Here we show that repression of Spry2 expression in TCL1-tg mouse and human B-cell lymphomas and cell lines is associated with dense DNA hypermethylation and was reversed by inhibition of DNA methylation. Spry2 expression was induced in normal splenic B cells by CD40/B-cell receptor costimulation and regulated a negative feedback loop that repressed MAPK-ERK signaling and decreased B-cell viability. Conversely, loss of Spry2 function hyperactivated MAPK-ERK signaling and caused increased B-cell proliferation. Combined, these results implicate epigenetic silencing of Spry2 expression in B lymphoma progression and suggest it as a companion lesion to ectopic TCL1 expression in enhancing MAPK-ERK pathway signaling.


Subject(s)
B-Lymphocytes/physiology , Cell Proliferation , Gene Expression Regulation, Neoplastic , Lymphoma, B-Cell/genetics , Membrane Proteins/physiology , Adaptor Proteins, Signal Transducing , Animals , B-Lymphocytes/metabolism , CD40 Antigens/metabolism , CD40 Antigens/physiology , DNA Methylation/physiology , Female , Gene Silencing/physiology , Humans , Intracellular Signaling Peptides and Proteins , MAP Kinase Signaling System/physiology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases , Tumor Cells, Cultured
13.
Cell ; 129(6): 1065-79, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17574021

ABSTRACT

The karyotypic chaos exhibited by human epithelial cancers complicates efforts to identify mutations critical for malignant transformation. Here we integrate complementary genomic approaches to identify human oncogenes. We show that activation of the ERK and phosphatidylinositol 3-kinase (PI3K) signaling pathways cooperate to transform human cells. Using a library of activated kinases, we identify several kinases that replace PI3K signaling and render cells tumorigenic. Whole genome structural analyses reveal that one of these kinases, IKBKE (IKKepsilon), is amplified and overexpressed in breast cancer cell lines and patient-derived tumors. Suppression of IKKepsilon expression in breast cancer cell lines that harbor IKBKE amplifications induces cell death. IKKepsilon activates the nuclear factor-kappaB (NF-kappaB) pathway in both cell lines and breast cancers. These observations suggest a mechanism for NF-kappaB activation in breast cancer, implicate the NF-kappaB pathway as a downstream mediator of PI3K, and provide a framework for integrated genomic approaches in oncogene discovery.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Genomics , I-kappa B Kinase/genetics , Alleles , Cell Line , Cell Transformation, Neoplastic , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Library , Genome , Humans , Models, Biological , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction
14.
Nanomedicine ; 3(1): 43-52, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17379168

ABSTRACT

Microtubules (MTs) are linked to cell mechanobiology. "Stable" and "dynamically unstable" microtubule (MT) subtypes are differentially sensitive to growth and distribution in serum starved (SS) versus full serum (FS) conditions. Atomic Force and Immunofluorescence microscopies were used to study the nanomechanical properties of the cell membrane in response to serum conditions and nocodazole. Nanomechanical properties of the cell membrane remain unchanged under SS/FS conditions even though there are drastic MT changes. The cell membrane is shown to depend on unstable MTs and the intermediate filament (IF) networks to maintain local stiffness. Measurements of local membrane nanomechanics in response to nocodazole display characteristic serum dependent decays. The responses suggest that the cell exists in a mechanical transition state. Stiffness is shown to depend on the interplay between dynamically unstable MTs, stable MTs and IFs which all act to impart a distinct cellular type of transient "metastability".


Subject(s)
Cell Membrane/metabolism , Fibroblasts/cytology , Microtubules/classification , Microtubules/metabolism , Actins/metabolism , Animals , Biomechanical Phenomena , Cell Death/drug effects , Cell Membrane/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Fibroblasts/drug effects , Kinetics , Mice , Microtubules/drug effects , Microtubules/ultrastructure , NIH 3T3 Cells , Nanoparticles , Nocodazole/pharmacology
15.
Blood ; 108(6): 1991-8, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16728701

ABSTRACT

Most lymphomas arise by transformation of germinal center (GC) B cells. TCL1, a proto-oncogene first recognized for its role in T-cell transformation, also induces GC B-cell malignancies when dysregulated in pEmu-B29-TCL1 transgenic (TCL1-tg) mice. Clonal B-cell lymphomas develop from polyclonal populations with latencies of 4 months or more, suggesting that secondary genetic events are required for full transformation. The goals of this study were to determine the GC-related effects of TCL1 dysregulation that contribute to tumor initiation and to identify companion genetic alterations in tumors that function in disease progression. We report that compared with wild-type (WT) cells, B cells from TCL1-tg mice activated in a manner resembling a T-dependent GC reaction show enhanced resistance to FAS-mediated apoptosis with CD40 stimulation, independent of a B-cell antigen receptor (BCR) rescue signal. Mitogenic stimulation of TCL1-tg B cells also resulted in increased expression of Aicda. These GC-related enhancements in survival and Aicda expression could underlie B-cell transformation. Supporting this notion, no B-cell lymphomas developed for 20 months when TCL1-tg mice were crossed onto an Oct coactivator from B cell (OCA-B)-deficient background to yield mice incapable of forming GCs. Spectral karyotype analyses showed that GC lymphomas from TCL1-tg mice exhibit recurrent chromosome translocations and trisomy 15, with corresponding MYC overexpression. We conclude that pEmu-B29-TCL1 transgenic B cells primed for transformation must experience the GC environment and, for at least some, develop genome instability to become fully malignant.


Subject(s)
B-Lymphocytes/metabolism , Cell Transformation, Neoplastic/genetics , Genomic Instability , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Animals , Apoptosis , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD40 Antigens/metabolism , Cell Differentiation , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Cytidine Deaminase/metabolism , Fas Ligand Protein , Female , Gene Expression , Genes, myc , Germinal Center/metabolism , Germinal Center/pathology , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Tumor Necrosis Factors/metabolism
16.
Blood ; 101(12): 5007-9, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12576313

ABSTRACT

Initially considered to be of natural killer (NK)-cell origin, CD4+ CD56+ blastic tumors (BTs) of skin have recently been proposed to be of dendritic cell lineage. We have previously described BTs with transformation to myelomonocytic leukemia. Here we report expression of the lymphoid proto-oncogene TCL1 in 10 (83%) of 12 BTs and in lymph node plasmacytoid dendritic cells (DC2s). TCL1 was also expressed in myelomonocytic blasts of 3 transformed BT cases but not in true NK-cell tumors (n = 18), de novo acute myelomonocytic leukemias (1 of 14, 7%), or mature T-cell malignancies (1 of 112, < 1%), with the exception of T-prolymphocytic leukemia (T-PLL). All BT cases were also positive for the DC2-associated marker CD123. These results further support derivation of BTs from DC2s, and demonstrate that TCL1 expression in this tumor is common to the immature blastoid, lymphoid-appearing, and subsequent myelomonocytic phases of this disease.


Subject(s)
CD4 Antigens/analysis , CD56 Antigen/analysis , Dendritic Cells/chemistry , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/analysis , Skin Neoplasms/chemistry , Bone Marrow/chemistry , Bone Marrow/immunology , Bone Marrow/pathology , Hematopoietic Stem Cells/chemistry , Hematopoietic Stem Cells/immunology , Humans , Immunohistochemistry , Immunophenotyping , Interleukin-3 Receptor alpha Subunit , Killer Cells, Natural , Leukemia, Myeloid/metabolism , Leukemia, Myelomonocytic, Acute/pathology , Leukemia, Prolymphocytic/metabolism , Leukemia, T-Cell/metabolism , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphoma/chemistry , Lymphoma, T-Cell/chemistry , Plasma Cells/chemistry , Plasma Cells/immunology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-akt , Receptors, Interleukin-3/analysis , Skin Neoplasms/immunology
17.
J Biol Chem ; 278(2): 948-55, 2003 Jan 10.
Article in English | MEDLINE | ID: mdl-12421830

ABSTRACT

Cis-regions and trans-factors controlling TCL1 oncogene expression are not known. We identified the functional TCL1 promoter by mapping four transcriptional start sites 24-30 bp downstream of a TATA box. A 424-bp fragment upstream of the major start site showed robust promoter activity comparable with SV40 in both TCL1 expressing and non-expressing cell lines. Additional constructs spanning 10 kb upstream and 20 kb downstream of the start site showed only modest increases in reporter activity indicating that TCL1 expression is primarily controlled by the promoter. Ten putative Sp1-binding sites were identified within 300 bp of the start site, and three of these specifically bound Sp1. A dose-dependent transactivation of the TCL1 promoter with Sp1 addition in Sp1-negative Drosophila SL2 cells was observed, and mutation of the three identified Sp1-binding sites significantly repressed reporter gene expression in 293T cells, confirming a key role for Sp1 in activating the TCL1 promoter in vivo. In TCL1 silent cell lines, CpG DNA methylation was rarely observed at functional Sp1 sites, and methylation of a previously reported NotI restriction site was associated with dense CpG methylation rather than endogenous TCL1 gene silencing. Together, these results indicate that Sp1 mediates transactivation of the TCL1 core promoter and that TCL1 gene silencing is not dependent on mechanisms involving Sp1 and NotI site methylation.


Subject(s)
Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Sp1 Transcription Factor/physiology , Transcriptional Activation , Base Sequence , Cell Line , CpG Islands , DNA Methylation , Humans , Molecular Sequence Data , Promoter Regions, Genetic
18.
Immunol Rev ; 187: 22-39, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12366680

ABSTRACT

Cell transplantation and tissue regeneration studies indicate a surprisingly broad developmental potential for lineage-committed hematopoietic stem cells (HSCs). Under these conditions HSCs transition into myocytes, neurons, hepatocytes or other types of nonhematopoietic effector cells. Equally impressive is the progression of committed neuronal stem cells (NSCs) to functional blood elements. Although critical cell-of-origin issues remain unresolved, the possibility of lineage switching is strengthened by a few well-controlled examples of cell-type conversion. At the molecular level, switching probably initiates from environmental signals that induce epigenetic modifications, resulting in changes in chromatin configuration. In turn, these changes affect patterns of gene expression that mediate divergent developmental programs. This review examines recent findings in nuclear reprogramming and cell fusion as potential causative mechanisms for transdifferentiation during normal and malignant hematopoiesis.


Subject(s)
Cell Differentiation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/pathology , Neoplasms/pathology , Animals , Cell Fusion , Cell Nucleus/metabolism , Gene Expression Regulation, Neoplastic , Germ Layers/cytology , Germ Layers/pathology , Hematopoietic Stem Cell Transplantation , Humans , Neurons/cytology , Neurons/pathology
19.
Biochemistry ; 41(20): 6376-82, 2002 May 21.
Article in English | MEDLINE | ID: mdl-12009899

ABSTRACT

AKT has a critical role in relaying cell survival and proliferation signals initiated by ligand binding to surface receptors in mammalian cells. Induction of AKT serine/threonine kinase activity is augmented by the T-cell leukemia-1 (TCL1) oncoprotein through a physical association requiring the AKT pleckstrin homology domain. Here, we used molecular modeling and identified an exposed hydrophobic patch composed of two discontinuous amino acid stretches near one end of the TCL1 beta-barrel that was required for a TCL1-AKT association. Site-directed mutations of this region did not affect TCL1 secondary structure, yet they disrupted interactions with AKT. This region was found in other members of the TCL1 oncoprotein family, such as TCL1b and MTCP1, and suggested a conserved, novel AKT binding domain. Interestingly, TCL1 and AKT co-localize in multiple cell compartments, but only extracts from the plasma membrane stimulate optimal complex formation in vitro. Identification of an AKT binding domain on TCL1 is an important step in deciphering the complex interactions that regulate AKT kinase activity in lymphocyte development and neoplasia within the immune system.


Subject(s)
Cytoplasm/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Intracellular Membranes/metabolism , Models, Molecular , Oncogene Proteins/chemistry , Oncogene Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cytoplasm/genetics , DNA-Binding Proteins/genetics , Group II Phospholipases A2 , Humans , Hydrophobic and Hydrophilic Interactions , Jurkat Cells , Leukemia, T-Cell/metabolism , Mice , Oncogene Proteins/genetics , Phospholipases A/metabolism , Protein Biosynthesis , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...