Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Biol Chem ; 291(3): 1307-19, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26631730

ABSTRACT

Aging involves progressive loss of cellular function and integrity, presumably caused by accumulated stochastic damage to cells. Alterations in energy metabolism contribute to aging, but how energy metabolism changes with age, how these changes affect aging, and whether they can be modified to modulate aging remain unclear. In locomotory muscle of post-fertile Caenorhabditis elegans, we identified a progressive decrease in cytosolic phosphoenolpyruvate carboxykinase (PEPCK-C), a longevity-associated metabolic enzyme, and a reciprocal increase in glycolytic pyruvate kinase (PK) that were necessary and sufficient to limit lifespan. Decline in PEPCK-C with age also led to loss of cellular function and integrity including muscle activity, and cellular senescence. Genetic and pharmacologic interventions of PEPCK-C, muscle activity, and AMPK signaling demonstrate that declines in PEPCK-C and muscle function with age interacted to limit reproductive life and lifespan via disrupted energy homeostasis. Quantifications of metabolic flux show that reciprocal changes in PEPCK-C and PK with age shunted energy metabolism toward glycolysis, reducing mitochondrial bioenergetics. Last, calorie restriction countered changes in PEPCK-C and PK with age to elicit anti-aging effects via TOR inhibition. Thus, a programmed metabolic event involving PEPCK-C and PK is a determinant of aging that can be modified to modulate aging.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Gene Expression Regulation, Developmental , Glycolysis , Mitochondrial Dynamics , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Pyruvate Kinase/metabolism , Aging , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/ultrastructure , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Caloric Restriction , Cytosol/enzymology , Cytosol/metabolism , Cytosol/ultrastructure , Energy Metabolism , Mutation , Phosphoenolpyruvate Carboxykinase (ATP)/antagonists & inhibitors , Phosphoenolpyruvate Carboxykinase (ATP)/genetics , Pyruvate Kinase/antagonists & inhibitors , Pyruvate Kinase/genetics , RNA Interference , Survival Analysis
2.
Neurodegener Dis ; 16(3-4): 179-83, 2016.
Article in English | MEDLINE | ID: mdl-26606044

ABSTRACT

BACKGROUND: Levodopa-induced dyskinesia (LID) is a disabling complication of levodopa therapy in Parkinson's disease (PD) with no effective treatments. Fluctuations in levels of levodopa constitute a key risk factor of LID. There is a pressing need for the development of a simple animal model of LID. Several genetic and toxin-based models of PD in Caenorhabditis elegans have been described, which have advanced our understanding of PD pathophysiology. We aimed to study levodopa-induced changes in a Parkinson's disease model of C. elegans expressing human α-synuclein. METHODS: We exposed the α-synuclein C. elegans to levodopa in continuous and alternating fashions. Automated behavioral analysis was then used to quantify changes in motor activity. Confocal microscopy was used next to quantify changes in dopamine receptor distribution and expression in motor neurons of live C. elegans. RESULTS: Chronic exposure to levodopa led to hyperactivity of the α-synuclein C. elegans without meaningful increase in motor activity. There was also an increase in peripheral clustering and expression of dopamine receptors in motor neurons. Both of these changes were significantly higher with alternating, compared to continuous, exposure to levodopa. CONCLUSIONS: This is the first report of changes in motor and dopamine receptors induced by levodopa in C. elegans overexpressing human α-synuclein. We propose that these phenotypes represent a simple animal model of LID in C. elegans. Such a model holds the promise of enabling high-throughput screenings for potential therapeutic targets and drug candidates.


Subject(s)
Antiparkinson Agents/toxicity , Dyskinesia, Drug-Induced/metabolism , Levodopa/toxicity , Parkinsonian Disorders/metabolism , Receptors, Dopamine/metabolism , alpha-Synuclein/metabolism , Actigraphy , Animals , Animals, Genetically Modified , Antiparkinson Agents/pharmacology , Caenorhabditis elegans , Dyskinesia, Drug-Induced/pathology , Humans , Levodopa/pharmacology , Microscopy, Confocal , Motor Activity/drug effects , Motor Activity/physiology , Motor Neurons/drug effects , Motor Neurons/metabolism , Motor Neurons/pathology , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/pathology , Pattern Recognition, Automated , alpha-Synuclein/genetics
3.
Oncogene ; 40(33): 5236-5246, 2021 08.
Article in English | MEDLINE | ID: mdl-34239044

ABSTRACT

Despite the fact that AML is the most common acute leukemia in adults, patient outcomes are poor necessitating the development of novel therapies. We identified that inhibition of Thioredoxin Reductase (TrxR) is a promising strategy for AML and report a highly potent and specific inhibitor of TrxR, S-250. Both pharmacologic and genetic inhibition of TrxR impairs the growth of human AML in mouse models. We found that TrxR inhibition leads to a rapid and marked impairment of metabolism in leukemic cells subsequently leading to cell death. TrxR was found to be a major and direct regulator of metabolism in AML cells through impacts on both glycolysis and the TCA cycle. Studies revealed that TrxR directly regulates GAPDH leading to a disruption of glycolysis and an increase in flux through the pentose phosphate pathway (PPP). The combined inhibition of TrxR and the PPP led to enhanced leukemia growth inhibition. Overall, TrxR abrogation, particularly with S-250, was identified as a promising strategy to disrupt AML metabolism.


Subject(s)
Pentose Phosphate Pathway , Thioredoxin-Disulfide Reductase , Cell Death , Citric Acid Cycle , Glycolysis , Humans
4.
Biomater Sci ; 8(14): 3935-3943, 2020 Jul 21.
Article in English | MEDLINE | ID: mdl-32662788

ABSTRACT

Non-Hodgkin's B cell lymphomas (NHL) include a diverse set of neoplasms that constitute ∼90% of all lymphomas and the largest subset of blood cancers. While chemotherapy is the first line of treatment, the efficacy of contemporary chemotherapies is hampered by dose-limiting toxicities. Partly due to suboptimal dosing, ∼40% of patients exhibit relapsed or refractory disease. Therefore more efficacious drug delivery systems are urgently needed to improve survival of NHL patients. In this study we demonstrate a new drug delivery platform for NHL based on the plant virus Potato virus X (PVX). We observed a binding affinity of PVX towards malignant B cells. In a metastatic mouse model of NHL, we show that systemically administered PVX home to tissues harboring malignant B cells. When loaded with the chemotherapy monomethyl auristatin (MMAE), the PVX nanocarrier enables effective delivery of MMAE to human B lymphoma cells in a NHL mouse model leading to inhibition of lymphoma growth in vivo and improved survival. Thus, PVX nanoparticle is a promising drug delivery platform for B cell malignancies.


Subject(s)
Antineoplastic Agents , Nanoparticles , Neoplasms , Potexvirus , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , B-Lymphocytes , Drug Delivery Systems , Humans , Neoplasms/drug therapy
5.
Front Immunol ; 11: 1941, 2020.
Article in English | MEDLINE | ID: mdl-32849651

ABSTRACT

Chimeric antigen receptor T cells (CAR-T cell) targeting CD19 are effective against several subtypes of CD19-expressing hematologic malignancies. Centralized manufacturing has allowed rapid expansion of this cellular therapy, but it may be associated with treatment delays due to the required logistics. We hypothesized that point of care manufacturing of CAR-T cells on the automated CliniMACS Prodigy® device allows reproducible and fast delivery of cells for the treatment of patients with non-Hodgkin lymphoma. Here we describe cell manufacturing results and characterize the phenotype and effector function of CAR-T cells used in a phase I/II study. We utilized a lentiviral vector delivering a second-generation CD19 CAR construct with 4-1BB costimulatory domain and TNFRSF19 transmembrane domain. Our data highlight the successful generation of CAR-T cells at numbers sufficient for all patients treated, a shortened duration of production from 12 to 8 days followed by fresh infusion into patients, and the detection of CAR-T cells in patient circulation up to 1-year post-infusion.


Subject(s)
Antigens, CD19/immunology , Cell Engineering , Immunotherapy, Adoptive , Lymphoma, Non-Hodgkin/therapy , Point-of-Care Systems , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/transplantation , Animals , Antigens, CD19/genetics , Antigens, CD19/metabolism , Automation , Cell Culture Techniques , Cells, Cultured , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Cytotoxicity, Immunologic , Humans , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/metabolism , Mice, Inbred NOD , Phenotype , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Autologous , Treatment Outcome , Workload , Xenograft Model Antitumor Assays
6.
Front Immunol ; 10: 812, 2019.
Article in English | MEDLINE | ID: mdl-31057545

ABSTRACT

Recognition of microorganism associated molecular patterns by epithelial cells elicits signaling cascades resulting in the production of host defense proteins. Lipocalin 24p3 is purported to be one such protein. 24p3 binds prokaryotic and eukaryotic siderophores and by sequestering iron laden bacterial siderophores it was believed to restrict bacterial replication. As such mice deficient for 24p3 are susceptible to systemic infections. However, it is not clear whether deficiency of 24p3 on the gut mucosa contributes to inflammation. In line with 24p3's function as a bacteriostat, it would be reasonable to assume that deficiencies in the control of intestinal flora from 24p3 absence play a role in inflammatory intestinal diseases. Surprisingly, we show 24p3 is a contributor of inflammation and 24p3 deficiency protects mice from dextran sodium sulfate (DSS)-induced colitis. 24p3 was found to be a negative regulator of platelet-derived growth factor (PDGF), which helps maintain the integrity of the gut mucosa. Neutralization of PDGF-BB abrogated resistance of 24p3 null mice to DSS confirming the direct link between 24p3 and PDGF-BB. Finally, iron handling in wild-type and 24p3-null mice upon DSS treatment also differed. In summary, differential iron levels and enhanced expression of PDGF-BB in 24p3 null mice confers resistance to DSS.


Subject(s)
Colitis/immunology , Lipocalin-2/immunology , Animals , Becaplermin/immunology , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Dextran Sulfate/toxicity , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Lipocalin-2/genetics , Mice , Mice, Knockout
7.
Sci Rep ; 9(1): 14916, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31624330

ABSTRACT

NK cell adoptive therapy is a promising cancer therapeutic approach, but there are significant challenges that limiting its feasibility and clinical efficacy. One difficulty is the paucity of clinical grade manufacturing platforms to support the large scale expansion of highly active NK cells. We created an NK cell feeder cell line termed 'NKF' through overexpressing membrane bound IL-21 that is capable of inducing robust and sustained proliferation (>10,000-fold expansion at 5 weeks) of highly cytotoxic NK cells. The expanded NK cells exhibit increased cytotoxic function against a panel of blood cancer and solid tumor cells as compared to IL-2-activated non-expanded NK cells. The NKF-expanded NK cells also demonstrate efficacy in mouse models of human sarcoma and T cell leukemia. Mechanistic studies revealed that membrane-bound IL-21 leads to an activation of a STAT3/c-Myc pathway and increased NK cell metabolism with a shift towards aerobic glycolysis. The NKF feeder cell line is a promising new platform that enables the large scale proliferation of highly active NK cells in support of large scale third party NK cell clinical studies that have been recently intiatied. These results also provide mechanistic insights into how membrane-bound IL-21 regulates NK cell expansion.


Subject(s)
Feeder Cells/metabolism , Immunotherapy/methods , Killer Cells, Natural/immunology , Neoplasms/therapy , Primary Cell Culture/methods , Animals , Cell Line, Tumor , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Proliferation , Coculture Techniques , Healthy Volunteers , Humans , Interleukins/immunology , Interleukins/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/transplantation , Mice , Neoplasms/immunology , Xenograft Model Antitumor Assays
8.
Elife ; 82019 02 13.
Article in English | MEDLINE | ID: mdl-30759065

ABSTRACT

Commonly-mutated genes have been found for many cancers, but less is known about mutations in cis-regulatory elements. We leverage gains in tumor-specific enhancer activity, coupled with allele-biased mutation detection from H3K27ac ChIP-seq data, to pinpoint potential enhancer-activating mutations in colorectal cancer (CRC). Analysis of a genetically-diverse cohort of CRC specimens revealed that microsatellite instable (MSI) samples have a high indel rate within active enhancers. Enhancers with indels show evidence of positive selection, increased target gene expression, and a subset is highly recurrent. The indels affect short homopolymer tracts of A/T and increase affinity for FOX transcription factors. We further demonstrate that signature mismatch-repair (MMR) mutations activate enhancers using a xenograft tumor metastasis model, where mutations are induced naturally via CRISPR/Cas9 inactivation of MLH1 prior to tumor cell injection. Our results suggest that MMR signature mutations activate enhancers in CRC tumor epigenomes to provide a selective advantage.


Subject(s)
Colorectal Neoplasms/genetics , DNA Mismatch Repair/genetics , Enhancer Elements, Genetic/genetics , Epigenome , Mutation/genetics , Acetylation , Animals , Base Sequence , Cell Line, Tumor , Gene Expression Regulation , Histones/metabolism , Humans , INDEL Mutation/genetics , Lysine/metabolism , Mice , Microsatellite Instability , Nucleotide Motifs/genetics , Phenotype , Selection, Genetic , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL