Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Cell ; 156(3): 590-602, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24485462

ABSTRACT

Therapy-resistant microenvironments represent a major barrier toward effective elimination of disseminated malignancies. Here, we show that select microenvironments can underlie resistance to antibody-based therapy. Using a humanized model of treatment refractory B cell leukemia, we find that infiltration of leukemia cells into the bone marrow rewires the tumor microenvironment to inhibit engulfment of antibody-targeted tumor cells. Resistance to macrophage-mediated killing can be overcome by combination regimens involving therapeutic antibodies and chemotherapy. Specifically, the nitrogen mustard cyclophosphamide induces an acute secretory activating phenotype (ASAP), releasing CCL4, IL8, VEGF, and TNFα from treated tumor cells. These factors induce macrophage infiltration and phagocytic activity in the bone marrow. Thus, the acute induction of stress-related cytokines can effectively target cancer cells for removal by the innate immune system. This synergistic chemoimmunotherapeutic regimen represents a potent strategy for using conventional anticancer agents to alter the tumor microenvironment and promote the efficacy of targeted therapeutics.


Subject(s)
Disease Models, Animal , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Tumor Microenvironment , Animals , Cyclophosphamide/therapeutic use , Cytokines/immunology , Drug Resistance, Neoplasm , Heterografts , Humans , Immunity, Innate , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Macrophages/immunology , Mice , Neoplasm Transplantation
2.
Cell ; 144(2): 296-309, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21241896

ABSTRACT

Though many individual transcription factors are known to regulate hematopoietic differentiation, major aspects of the global architecture of hematopoiesis remain unknown. Here, we profiled gene expression in 38 distinct purified populations of human hematopoietic cells and used probabilistic models of gene expression and analysis of cis-elements in gene promoters to decipher the general organization of their regulatory circuitry. We identified modules of highly coexpressed genes, some of which are restricted to a single lineage but most of which are expressed at variable levels across multiple lineages. We found densely interconnected cis-regulatory circuits and a large number of transcription factors that are differentially expressed across hematopoietic states. These findings suggest a more complex regulatory system for hematopoiesis than previously assumed.


Subject(s)
Gene Expression Regulation , Gene Regulatory Networks , Hematopoiesis , Transcription Factors/metabolism , Gene Expression Profiling , Humans
3.
Blood ; 142(3): 260-273, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37192303

ABSTRACT

Although treatment of multiple myeloma (MM) with daratumumab significantly extends the patient's lifespan, resistance to therapy is inevitable. ISB 1342 was designed to target MM cells from patients with relapsed/refractory MM (r/r MM) displaying lower sensitivity to daratumumab. ISB 1342 is a bispecific antibody with a high-affinity Fab binding to CD38 on tumor cells on a different epitope than daratumumab and a detuned scFv domain affinity binding to CD3ε on T cells, to mitigate the risk of life-threatening cytokine release syndrome, using the Bispecific Engagement by Antibodies based on the TCR (BEAT) platform. In vitro, ISB 1342 efficiently killed cell lines with different levels of CD38, including those with a lower sensitivity to daratumumab. In a killing assay where multiple modes of action were enabled, ISB 1342 showed higher cytotoxicity toward MM cells compared with daratumumab. This activity was retained when used in sequential or concomitant combinations with daratumumab. The efficacy of ISB 1342 was maintained in daratumumab-treated bone marrow patient samples showing lower sensitivity to daratumumab. ISB 1342 induced complete tumor control in 2 therapeutic mouse models, unlike daratumumab. Finally, in cynomolgus monkeys, ISB 1342 displayed an acceptable toxicology profile. These data suggest that ISB 1342 may be an option in patients with r/r MM refractory to prior anti-CD38 bivalent monoclonal antibody therapies. It is currently being developed in a phase 1 clinical study.


Subject(s)
Antibodies, Bispecific , Multiple Myeloma , Animals , Mice , ADP-ribosyl Cyclase 1/metabolism , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Multiple Myeloma/drug therapy , T-Lymphocytes/pathology
4.
Genes Dev ; 30(3): 321-36, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26833731

ABSTRACT

Oncogene-induced senescence (OIS) and therapy-induced senescence (TIS), while tumor-suppressive, also promote procarcinogenic effects by activating the DNA damage response (DDR), which in turn induces inflammation. This inflammatory response prominently includes an array of cytokines known as the senescence-associated secretory phenotype (SASP). Previous observations link the transcription-associated methyltransferase and oncoprotein MLL1 to the DDR, leading us to investigate the role of MLL1 in SASP expression. Our findings reveal direct MLL1 epigenetic control over proproliferative cell cycle genes: MLL1 inhibition represses expression of proproliferative cell cycle regulators required for DNA replication and DDR activation, thus disabling SASP expression. Strikingly, however, these effects of MLL1 inhibition on SASP gene expression do not impair OIS and, furthermore, abolish the ability of the SASP to enhance cancer cell proliferation. More broadly, MLL1 inhibition also reduces "SASP-like" inflammatory gene expression from cancer cells in vitro and in vivo independently of senescence. Taken together, these data demonstrate that MLL1 inhibition may be a powerful and effective strategy for inducing cancerous growth arrest through the direct epigenetic regulation of proliferation-promoting genes and the avoidance of deleterious OIS- or TIS-related tumor secretomes, which can promote both drug resistance and tumor progression.


Subject(s)
Cellular Senescence/genetics , Gene Expression Regulation, Neoplastic/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Signal Transduction/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Line , Cell Proliferation , DNA Damage , Gene Knockdown Techniques , HEK293 Cells , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Humans , Inflammation/genetics , MCF-7 Cells , Myeloid-Lymphoid Leukemia Protein/antagonists & inhibitors , NF-kappa B/metabolism , Neoplasms/physiopathology , Phenotype
5.
Nature ; 546(7658): 381-386, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28562591

ABSTRACT

Metabolic production of acetyl coenzyme A (acetyl-CoA) is linked to histone acetylation and gene regulation, but the precise mechanisms of this process are largely unknown. Here we show that the metabolic enzyme acetyl-CoA synthetase 2 (ACSS2) directly regulates histone acetylation in neurons and spatial memory in mammals. In a neuronal cell culture model, ACSS2 increases in the nuclei of differentiating neurons and localizes to upregulated neuronal genes near sites of elevated histone acetylation. A decrease in ACSS2 lowers nuclear acetyl-CoA levels, histone acetylation, and responsive expression of the cohort of neuronal genes. In adult mice, attenuation of hippocampal ACSS2 expression impairs long-term spatial memory, a cognitive process that relies on histone acetylation. A decrease in ACSS2 in the hippocampus also leads to defective upregulation of memory-related neuronal genes that are pre-bound by ACSS2. These results reveal a connection between cellular metabolism, gene regulation, and neural plasticity and establish a link between acetyl-CoA generation 'on-site' at chromatin for histone acetylation and the transcription of key neuronal genes.


Subject(s)
Acetate-CoA Ligase/metabolism , Hippocampus/enzymology , Hippocampus/physiology , Histones/metabolism , Memory/physiology , Neuronal Plasticity/genetics , Transcriptional Activation , Acetate-CoA Ligase/deficiency , Acetate-CoA Ligase/genetics , Acetyl Coenzyme A/metabolism , Acetylation , Animals , Cell Differentiation , Cell Nucleus/metabolism , Cells, Cultured , Chromatin/enzymology , Chromatin/genetics , Chromatin/metabolism , Gene Expression Regulation, Enzymologic , Hippocampus/metabolism , Histones/chemistry , Memory Consolidation/physiology , Mice , Neuronal Plasticity/physiology , Neurons/cytology , Neurons/metabolism , Up-Regulation
6.
J Pharmacol Exp Ther ; 380(3): 210-219, 2022 03.
Article in English | MEDLINE | ID: mdl-35031585

ABSTRACT

Etavopivat is an investigational, oral, small molecule activator of erythrocyte pyruvate kinase (PKR) in development for the treatment of sickle cell disease (SCD) and other hemoglobinopathies. PKR activation is proposed to ameliorate the sickling of SCD red blood cells (RBCs) through multiple mechanisms, including reduction of 2,3-diphosphoglycerate (2,3-DPG), which consequently increases hemoglobin (Hb)-oxygen affinity; increased binding of oxygen reduces sickle hemoglobin polymerization and sickling. In addition, PKR activation increases adenosine triphosphate (ATP) produced via glycolytic flux, which helps preserve membrane integrity and RBC deformability. We evaluated the pharmacodynamic response to etavopivat in nonhuman primates (NHPs) and in healthy human subjects and evaluated the effects in RBCs from patients with SCD after ex vivo treatment with etavopivat. A single dose of etavopivat decreased 2,3-DPG in NHPs and healthy subjects. Hb-oxygen affinity was significantly increased in healthy subjects after 24 hours. After daily dosing of etavopivat over 5 consecutive days in NHPs, ATP was increased by 38% from baseline. Etavopivat increased Hb-oxygen affinity and reduced sickling in RBCs collected from patients with SCD with either homozygous hemoglobin S or hemoglobin S and C disease. Collectively, these results demonstrate the ability of etavopivat to decrease 2,3-DPG and increase ATP, resulting in increased Hb-oxygen affinity and improved sickle RBC function. Etavopivat is currently being evaluated in clinical trials for the treatment of SCD. SIGNIFICANCE STATEMENT: Etavopivat, a small molecule activator of the glycolytic enzyme erythrocyte pyruvate kinase, decreased 2,3-diphosphoglycerate in red blood cells (RBCs) from nonhuman primates and healthy subjects and significantly increased hemoglobin (Hb)-oxygen affinity in healthy subjects. Using ex vivo RBCs from donors with sickle cell disease (SCD) (homozygous hemoglobin S or hemoglobin S and C genotype), etavopivat increased Hb-oxygen affinity and reduced sickling under deoxygenation. Etavopivat shows promise as a treatment for SCD that could potentially reduce vaso-occlusion and improve anemia.


Subject(s)
Anemia, Sickle Cell , Hemoglobin, Sickle , 2,3-Diphosphoglycerate/metabolism , 2,3-Diphosphoglycerate/pharmacology , Adenosine Triphosphate/metabolism , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/metabolism , Animals , Erythrocytes/metabolism , Hemoglobin, Sickle/metabolism , Hemoglobin, Sickle/pharmacology , Hemoglobin, Sickle/therapeutic use , Hemoglobins/metabolism , Humans , Oxygen/metabolism , Pyruvate Kinase/metabolism , Pyruvate Kinase/pharmacology , Pyruvate Kinase/therapeutic use , Pyruvic Acid/pharmacology
7.
J Immunol ; 202(6): 1885-1894, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30710044

ABSTRACT

Development of targeted cancer therapy requires a thorough understanding of mechanisms of tumorigenesis as well as mechanisms of action of therapeutics. This is challenging because by the time patients are diagnosed with cancer, early events of tumorigenesis have already taken place. Similarly, development of cancer immunotherapies is hampered by a lack of appropriate small animal models with autologous human tumor and immune system. In this article, we report the development of a mouse model of human acute myeloid leukemia (AML) with autologous immune system for studying early events of human leukemogenesis and testing the efficacy of immunotherapeutics. To develop such a model, human hematopoietic stem/progenitor cells (HSPC) are transduced with lentiviruses expressing a mutated form of nucleophosmin (NPM1), referred to as NPM1c. Following engraftment into immunodeficient mice, transduced HSPCs give rise to human myeloid leukemia, whereas untransduced HSPCs give rise to human immune cells in the same mice. The de novo AML, with CD123+ leukemic stem or initiating cells (LSC), resembles NPM1c+ AML from patients. Transcriptional analysis of LSC and leukemic cells confirms similarity of the de novo leukemia generated in mice with patient leukemia and suggests Myc as a co-operating factor in NPM1c-driven leukemogenesis. We show that a bispecific conjugate that binds both CD3 and CD123 eliminates CD123+ LSCs in a T cell-dependent manner both in vivo and in vitro. These results demonstrate the utility of the NPM1c+ AML model with an autologous immune system for studying early events of human leukemogenesis and for evaluating efficacy and mechanism of immunotherapeutics.


Subject(s)
Carcinogenesis , Leukemia, Myeloid , Nuclear Proteins , Xenograft Model Antitumor Assays , Animals , Hematopoietic Stem Cells , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Nucleophosmin
8.
Nature ; 527(7576): 105-9, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26524528

ABSTRACT

Macroautophagy (hereafter referred to as autophagy) is a catabolic membrane trafficking process that degrades a variety of cellular constituents and is associated with human diseases. Although extensive studies have focused on autophagic turnover of cytoplasmic materials, little is known about the role of autophagy in degrading nuclear components. Here we report that the autophagy machinery mediates degradation of nuclear lamina components in mammals. The autophagy protein LC3/Atg8, which is involved in autophagy membrane trafficking and substrate delivery, is present in the nucleus and directly interacts with the nuclear lamina protein lamin B1, and binds to lamin-associated domains on chromatin. This LC3-lamin B1 interaction does not downregulate lamin B1 during starvation, but mediates its degradation upon oncogenic insults, such as by activated RAS. Lamin B1 degradation is achieved by nucleus-to-cytoplasm transport that delivers lamin B1 to the lysosome. Inhibiting autophagy or the LC3-lamin B1 interaction prevents activated RAS-induced lamin B1 loss and attenuates oncogene-induced senescence in primary human cells. Our study suggests that this new function of autophagy acts as a guarding mechanism protecting cells from tumorigenesis.


Subject(s)
Autophagy , Nuclear Lamina/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autophagy-Related Protein 8 Family , Cell Transformation, Neoplastic , Cells, Cultured , Cellular Senescence , Chromatin/chemistry , Chromatin/metabolism , Cytoplasm/metabolism , Fibroblasts , HEK293 Cells , Humans , Lamin Type B/genetics , Lamin Type B/metabolism , Lysosomes/metabolism , Mice , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Oncogene Protein p21(ras)/metabolism , Protein Binding , Proteolysis
9.
BMC Cancer ; 19(1): 832, 2019 Aug 23.
Article in English | MEDLINE | ID: mdl-31443703

ABSTRACT

BACKGROUND: Blood-based methods using cell-free DNA (cfDNA) are under development as an alternative to existing screening tests. However, early-stage detection of cancer using tumor-derived cfDNA has proven challenging because of the small proportion of cfDNA derived from tumor tissue in early-stage disease. A machine learning approach to discover signatures in cfDNA, potentially reflective of both tumor and non-tumor contributions, may represent a promising direction for the early detection of cancer. METHODS: Whole-genome sequencing was performed on cfDNA extracted from plasma samples (N = 546 colorectal cancer and 271 non-cancer controls). Reads aligning to protein-coding gene bodies were extracted, and read counts were normalized. cfDNA tumor fraction was estimated using IchorCNA. Machine learning models were trained using k-fold cross-validation and confounder-based cross-validations to assess generalization performance. RESULTS: In a colorectal cancer cohort heavily weighted towards early-stage cancer (80% stage I/II), we achieved a mean AUC of 0.92 (95% CI 0.91-0.93) with a mean sensitivity of 85% (95% CI 83-86%) at 85% specificity. Sensitivity generally increased with tumor stage and increasing tumor fraction. Stratification by age, sequencing batch, and institution demonstrated the impact of these confounders and provided a more accurate assessment of generalization performance. CONCLUSIONS: A machine learning approach using cfDNA achieved high sensitivity and specificity in a large, predominantly early-stage, colorectal cancer cohort. The possibility of systematic technical and institution-specific biases warrants similar confounder analyses in other studies. Prospective validation of this machine learning method and evaluation of a multi-analyte approach are underway.


Subject(s)
Biomarkers, Tumor , Circulating Tumor DNA , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Genome, Human , Genomics , Machine Learning , Aged , Aged, 80 and over , Colorectal Neoplasms/blood , Computational Biology/methods , Female , Gene Expression Profiling , Genomics/methods , Humans , Male , Middle Aged , Neoplasm Staging , ROC Curve , Reproducibility of Results , Transcriptome
10.
Genome Res ; 25(2): 179-88, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25391375

ABSTRACT

Despite overwhelming evidence that transcriptional activation by TP53 is critical for its tumor suppressive activity, the mechanisms by which TP53 engages the genome in the context of chromatin to activate transcription are not well understood. Using a compendium of novel and existing genome-wide data sets, we examined the relationship between TP53 binding and the dynamics of the local chromatin environment. Our analysis revealed three distinct categories of TP53 binding events that differ based on the dynamics of the local chromatin environment. The first class of TP53 binding events occurs near transcriptional start sites (TSS) and is defined by previously characterized promoter-associated chromatin modifications. The second class comprises a large cohort of preestablished, promoter-distal enhancer elements that demonstrates dynamic histone acetylation and transcription upon TP53 binding. The third class of TP53 binding sites is devoid of classic chromatin modifications and, remarkably, fall within regions of inaccessible chromatin, suggesting that TP53 has intrinsic pioneer factor activity and binds within structurally inaccessible regions of chromatin. Intriguingly, these inaccessible TP53 binding sites feature several enhancer-like properties in cell types within the epithelial lineage, indicating that TP53 binding events include a group of "proto-enhancers" that become active enhancers given the appropriate cellular context. These data indicate that TP53, along with TP63, may act as pioneer factors to specify epithelial enhancers. Further, these findings suggest that rather than following a global cell-type invariant stress response program, TP53 may tune its response based on the lineage-specific epigenomic landscape.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , Genome, Human , Tumor Suppressor Protein p53/metabolism , Acetylation , Binding Sites , Computational Biology , Databases, Genetic , Enhancer Elements, Genetic , Epigenesis, Genetic , Gene Expression Regulation , Histones/metabolism , Humans , Methylation , Organ Specificity , Protein Binding , Transcription Factors/metabolism , Transcription Initiation Site , Transcriptional Activation
11.
Blood Adv ; 5(9): 2385-2390, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33944896

ABSTRACT

Sickle cell anemia (SCA) results from an abnormal sickle hemoglobin (HbS). HbS polymerizes upon deoxygenation, resulting in red blood cell (RBC) sickling and membrane damage that cause vaso-occlusions and hemolysis. Sickle RBCs contain less adenosine triphosphate and more 2,3-diphosphoglycerate than normal RBCs, which allosterically reduces hemoglobin (Hb) oxygen (O2) affinity (ie, increases the partial pressure of oxygen at which hemoglobin is 50% saturated with oxygen [P50]), potentiating HbS polymerization. Herein, we tested the effect of investigational agent FT-4202, an RBC pyruvate kinase (PKR) activator, on RBC sickling and membrane damage by administering it to Berkeley SCA mice. Two-week oral FT-4202 administration was well tolerated, decreasing HbS P50 to levels similar to HbA and demonstrating beneficial biological effects. In FT-4202-treated animals, there was reduced sickling in vivo, demonstrated by fewer irreversibly sickled cells, and improved RBC deformability, assessed at varying shear stress. Controlled deoxygenation followed by reoxygenation of RBCs obtained from the blood of FT-4202-treated mice showed a shift in the point of sickling to a lower partial pressure of oxygen (pO2). This led to a nearly 30% increase in RBC survival and a 1.7g/dL increase in Hb level in the FT-4202-treated SCA mice. Overall, our results in SCA mice suggest that FT-4202 might be a potentially useful oral antisickling agent that warrants investigation in patients with SCA.


Subject(s)
Anemia, Sickle Cell , Hemoglobin, Sickle , Anemia, Sickle Cell/drug therapy , Animals , Antisickling Agents , Erythrocytes, Abnormal , Humans , Mice , Pyruvate Kinase
12.
PLoS One ; 11(11): e0166280, 2016.
Article in English | MEDLINE | ID: mdl-27855183

ABSTRACT

T lymphocytes require signals from self-peptides and cytokines, most notably interleukins 7 and 15 (IL-7, IL-15), for survival. While mouse T cells die rapidly if IL-7 or IL-15 is withdrawn, human T cells can survive prolonged withdrawal of IL-7 and IL-15. Here we show that IL-7 and IL-15 are required to maintain human T cell proliferative capacity through the STAT5 signaling pathway. T cells from humanized mice proliferate better if stimulated in the presence of human IL-7 or IL-15 or if T cells are exposed to human IL-7 or IL-15 in mice. Freshly isolated T cells from human peripheral blood lose proliferative capacity if cultured for 24 hours in the absence of IL-7 or IL-15. We further show that phosphorylation of STAT5 correlates with proliferation and inhibition of STAT5 reduces proliferation. These results reveal a novel role of IL-7 and IL-15 in maintaining human T cell function, provide an explanation for T cell dysfunction in humanized mice, and have significant implications for in vitro studies with human T cells.


Subject(s)
Interleukin-15/metabolism , Interleukin-7/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Animals , Cell Proliferation , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation , Lymphocyte Count , Mice , Phosphorylation , Up-Regulation
13.
Science ; 354(6316): 1160-1165, 2016 12 02.
Article in English | MEDLINE | ID: mdl-27789795

ABSTRACT

Blocking Programmed Death-1 (PD-1) can reinvigorate exhausted CD8 T cells (TEX) and improve control of chronic infections and cancer. However, whether blocking PD-1 can reprogram TEX into durable memory T cells (TMEM) is unclear. We found that reinvigoration of TEX in mice by PD-L1 blockade caused minimal memory development. After blockade, reinvigorated TEX became reexhausted if antigen concentration remained high and failed to become TMEM upon antigen clearance. TEX acquired an epigenetic profile distinct from that of effector T cells (TEFF) and TMEM cells that was minimally remodeled after PD-L1 blockade. This finding suggests that TEX are a distinct lineage of CD8 T cells. Nevertheless, PD-1 pathway blockade resulted in transcriptional rewiring and reengagement of effector circuitry in the TEX epigenetic landscape. These data indicate that epigenetic fate inflexibility may limit current immunotherapies.


Subject(s)
B7-H1 Antigen/genetics , CD8-Positive T-Lymphocytes/immunology , Cellular Reprogramming/genetics , Epigenesis, Genetic , Immunologic Memory/genetics , Animals , B7-H1 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/transplantation , Cell Lineage/genetics , Cellular Reprogramming/immunology , Female , Gene Regulatory Networks , Immunotherapy , Interleukin-7/metabolism , Mice , Mice, Inbred C57BL , Transcription, Genetic
14.
DNA Repair (Amst) ; 3(1): 33-41, 2004 Jan 05.
Article in English | MEDLINE | ID: mdl-14697757

ABSTRACT

Cellular life depends upon the preservation and transmission of genetic material. Double stranded DNA breaks (DSBs) cause catastrophic gene loss in cell division and must be promptly and accurately repaired. In eukaryotes DSBs may be repaired by either non-homologous end-joining (NHEJ), single strand annealing or homologous recombination (HR). Vertebrate NHEJ has been shown to depend upon the DNA-dependent protein kinase (DNA-PK) consisting of the phosphatidylinositol 3 (PI 3)-kinase like (PIKK) catalytic sub-unit (DNA-PKcs) and the DNA targeting factor Ku. Our analysis of recently completed genomes found several novel PIKKs in Anopheles gambiae and Drosophila melanogaster including a novel mosquito DNA-PKcs orthologue, the first non-vertebrate DNA-PKcs described to date. We also detected a DNA-PKcs fragment in the high quality EST set of Apis mellifera ligustica (honey bee) suggesting that DNA-PK is a far older and more important eukaryotic complex than previously thought.


Subject(s)
Anopheles/enzymology , Bees/enzymology , Culicidae/enzymology , Drosophila melanogaster/enzymology , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Anopheles/classification , Anopheles/genetics , Antigens, Nuclear/metabolism , Arthropods/enzymology , Bees/genetics , Culicidae/genetics , DNA-Activated Protein Kinase , DNA-Binding Proteins/metabolism , Drosophila melanogaster/genetics , Humans , Ku Autoantigen , Molecular Sequence Data , Nuclear Proteins , Protein Serine-Threonine Kinases/genetics , Sequence Homology, Amino Acid , Vertebrates/genetics , Vertebrates/metabolism
15.
Cancer Immunol Res ; 1: 217-222, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24466562

ABSTRACT

The 12th annual summer symposium of The Koch Institute for Integrative Cancer Research at MIT was held in Cambridge, MA, on June 14th, 1023. The symposium entitled "Cancer Immunology and Immunotherapy" focused on recent advances in preclinical research in basic immunology and biomedical engineering, and their clinical application in cancer therapies. The day-long gathering also provided a forum for discussion and potential collaborations between engineers and clinical investigators. The major topics presented include: (i) enhancement of adoptive cell therapy by engineering to improve the ability and functionality of T-cells against tumor cells; (ii) current therapies using protein and antibody therapeutics to modulate endogenous anti-tumor immunity; and (iii) new technologies to identify molecular targets and assess therapeutic efficacy, and devices to control and target drug delivery more effectively and efficiently.

16.
Oncogene ; 32(8): 1066-1072, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-22484426

ABSTRACT

Although numerous mouse models of B-cell malignancy have been developed via the enforced expression of defined oncogenic lesions, the feasibility of generating lineage-defined human B-cell malignancies using mice reconstituted with modified human hematopoietic stem cells (HSCs) remains unclear. In fact, whether human cells can be transformed as readily as murine cells by simple oncogene combinations is a subject of considerable debate. Here, we describe the development of humanized mouse model of MYC/BCL2-driven 'double-hit' lymphoma. By engrafting human HSCs transduced with the oncogene combination into immunodeficient mice, we generate a fatal B malignancy with complete penetrance. This humanized-MYC/BCL2-model (hMB) accurately recapitulates the histopathological and clinical aspects of steroid-, chemotherapy- and rituximab-resistant human 'double-hit' lymphomas that involve the MYC and BCL2 loci. Notably, this model can serve as a platform for the evaluation of antibody-based therapeutics. As a proof of principle, we used this model to show that the anti-CD52 antibody alemtuzumab effectively eliminates lymphoma cells from the spleen, liver and peripheral blood, but not from the brain. The hMB humanized mouse model underscores the synergy of MYC and BCL2 in 'double-hit' lymphomas in human patients. Additionally, our findings highlight the utility of humanized mouse models in interrogating therapeutic approaches, particularly human-specific monoclonal antibodies.


Subject(s)
Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-myc/biosynthesis , Animals , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Lymphoma, B-Cell/genetics , Mice , Penetrance , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Survival Analysis
17.
Cell Mol Immunol ; 9(3): 215-24, 2012 May.
Article in English | MEDLINE | ID: mdl-22425741

ABSTRACT

Humanized mice are immunodeficient animals engrafted with human hematopoietic stem cells that give rise to various lineages of human blood cells throughout the life of the mouse. This article reviews recent advances in the generation of humanized mice, focusing on practical considerations. We discuss features of different immunodeficient recipient mouse strains, sources of human hematopoietic stem cells, advances in expansion and genetic modification of hematopoietic stem cells, and techniques to modulate the cytokine environment of recipient mice, in order to enhance reconstitution of specific human blood lineage cells. We highlight the opportunities created by new technologies and discuss practical considerations on how to make best use of the widening array of basic models for specific research applications.


Subject(s)
Cell Survival , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Animals , Cytokines/immunology , Cytokines/metabolism , Genetic Engineering , Hematopoiesis/genetics , Hematopoiesis/immunology , Humans , Immunomodulation , Mice , Mice, Mutant Strains , Mice, SCID , Research Design , Stem Cell Niche , Transplantation, Heterologous
18.
ACS Nano ; 6(1): 81-8, 2012 Jan 24.
Article in English | MEDLINE | ID: mdl-22176729

ABSTRACT

The ability to control the timing and order of release of different therapeutic drugs will play a pivotal role in improving patient care and simplifying treatment regimes in the clinic. The controlled sequential release of a broad range of small and macromolecules from thin film coatings offers a simple way to provide complex localized dosing in vivo. Here we show that it is possible to take advantage of the structure of certain nanomaterials to control release regimes from a scale of hours to months. Graphene oxide (GO) is a two-dimensional charged nanomaterial that can be used to create barrier layers in multilayer thin films, trapping molecules of interest for controlled release. Protein-loaded polyelectrolyte multilayer films were fabricated using layer-by-layer assembly incorporating a hydrolytically degradable cationic poly(ß-amino ester) (Poly1) with a model protein antigen, ovalbumin (ova), in a bilayer architecture along with positively and negatively functionalized GO capping layers for the degradable protein films. Ova release without the GO layers takes place in less than 1 h but can be tuned to release from 30 to 90 days by varying the number of bilayers of functionalized GO in the multilayer architecture. We demonstrate that proteins can be released in sequence with multi-day gaps between the release of each species by incorporating GO layers between protein loaded layers. In vitro toxicity assays of the individual materials on proliferating hematopoietic stem cells (HSCs) indicated limited cytotoxic effects with HSCs able to survive for the full 10 days of normal culture in the presence of Poly1 and the GO sheets. This approach provides a new route for storage of therapeutics in a solid-state thin film for subsequent delivery in a time-controlled and sequential fashion.


Subject(s)
Delayed-Action Preparations/administration & dosage , Graphite/chemistry , Hematopoietic Stem Cells/drug effects , Nanocapsules/chemistry , Proteins/administration & dosage , Cell Survival/drug effects , Cells, Cultured , Diffusion , Hematopoietic Stem Cells/cytology , Humans , Materials Testing , Nanocapsules/administration & dosage , Proteins/chemistry
19.
Cell Mol Immunol ; 8(5): 415-23, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21666707

ABSTRACT

How tumor-infiltrating lymphocytes (TILs) that are tumor-specific but functionally tolerant persist in the antigen-expressing tumor tissue is largely unknown. We have previously developed a modified TRansgenic Adenocarcinoma of the Mouse Prostate (TRAMP) model where prostate cancer cells express the T-cell epitope SIYRYYGL (SIY) recognized by CD8 T cells expressing the 2C T-cell receptor (TCR) (referred to as TRP-SIY mice). In TRP-SIY mice, activated 2C T cells rapidly become tolerant following infiltration into the prostate tumor. In this study, we show that tolerant 2C T cells persist in the prostate tumor of TRP-SIY mice by proliferating slowly in a tumor-dependent, but antigen-, interleukin (IL)-7- and IL-15-independent manner. We also show that disappearance of 2C T cells from the lymphoid organs of TRP-SIY mice are due to antigen-induced T-cell contraction rather than altered trafficking or generalized T-cell depletion in the mice. Finally, we show that clonal T cells unreactive to SIY are equally capable of persisting in the prostate tumor. These findings suggest that while functional tolerance of TILs is induced by antigen, persistence of tolerant TILs in the tumor tissue is mediated by a novel mechanism: slow proliferation independent of antigen and homeostatic cytokines. These results also allow CD8 T-cell survival in the tumor environment to be compared with T-cell survival in chronic infection.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Immune Tolerance , Lymphocytes, Tumor-Infiltrating/immunology , Prostate/immunology , Prostatic Neoplasms/immunology , Adoptive Transfer , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , Flow Cytometry , Humans , Influenza A Virus, H1N1 Subtype , Interleukin-15/immunology , Interleukin-7/immunology , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/pathology , Real-Time Polymerase Chain Reaction , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Transduction, Genetic
20.
PLoS One ; 6(4): e18382, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21559522

ABSTRACT

Increasing demand for human hematopoietic stem cells (HSCs) in clinical and research applications necessitates expansion of HSCs in vitro. Before these cells can be used they must be carefully evaluated to assess their stem cell activity. Here, we expanded cord blood CD34(+) CD133(+) cells in a defined medium containing angiopoietin like 5 and insulin-like growth factor binding protein 2 and evaluated the cells for stem cell activity in NOD-SCID Il2rg(-/-) (NSG) mice by multi-lineage engraftment, long term reconstitution, limiting dilution and serial reconstitution. The phenotype of expanded cells was characterized by flow cytometry during the course of expansion and following engraftment in mice. We show that the SCID repopulating activity resides in the CD34(+) CD133(+) fraction of expanded cells and that CD34(+) CD133(+) cell number correlates with SCID repopulating activity before and after culture. The expanded cells mediate long-term hematopoiesis and serial reconstitution in NSG mice. Furthermore, they efficiently reconstitute not only neonate but also adult NSG recipients, generating human blood cell populations similar to those reported in mice reconstituted with uncultured human HSCs. These findings suggest an expansion of long term HSCs in our culture and show that expression of CD34 and CD133 serves as a marker for HSC activity in human cord blood cell cultures. The ability to expand human HSCs in vitro should facilitate clinical use of HSCs and large-scale construction of humanized mice from the same donor for research applications.


Subject(s)
Angiopoietins/metabolism , Antigens, CD34/biosynthesis , Antigens, CD/biosynthesis , Glycoproteins/biosynthesis , Interleukin Receptor Common gamma Subunit/genetics , AC133 Antigen , Angiopoietin-like Proteins , Animals , Blood Platelets/metabolism , Fetal Blood/metabolism , Flow Cytometry/methods , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Peptides
SELECTION OF CITATIONS
SEARCH DETAIL