Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 298(9): 102336, 2022 09.
Article in English | MEDLINE | ID: mdl-35931111

ABSTRACT

Mitochondrial chelatable iron contributes to the severity of several injury processes, including ischemia/reperfusion, oxidative stress, and drug toxicity. However, methods to measure this species in living cells are lacking. To measure mitochondrial chelatable iron in living cells, here we synthesized a new fluorescent indicator, mitoferrofluor (MFF). We designed cationic MFF to accumulate electrophoretically in polarized mitochondria, where a reactive group then forms covalent adducts with mitochondrial proteins to retain MFF even after subsequent depolarization. We also show in cell-free medium that Fe2+ (and Cu2+), but not Fe3+, Ca2+, or other biologically relevant divalent cations, strongly quenched MFF fluorescence. Using confocal microscopy, we demonstrate in hepatocytes that red MFF fluorescence colocalized with the green fluorescence of the mitochondrial membrane potential (ΔΨm) indicator, rhodamine 123 (Rh123), indicating selective accumulation into the mitochondria. Unlike Rh123, mitochondria retained MFF after ΔΨm collapse. Furthermore, intracellular delivery of iron with membrane-permeant Fe3+/8-hydroxyquinoline (FeHQ) quenched MFF fluorescence by ∼80% in hepatocytes and other cell lines, which was substantially restored by the membrane-permeant transition metal chelator pyridoxal isonicotinoyl hydrazone. We also show FeHQ quenched the fluorescence of cytosolically coloaded calcein, another Fe2+ indicator, confirming that Fe3+ in FeHQ undergoes intracellular reduction to Fe2+. Finally, MFF fluorescence did not change after addition of the calcium mobilizer thapsigargin, which shows MFF is insensitive to physiologically relevant increases of mitochondrial Ca2+. In conclusion, the new sensor reagent MFF fluorescence is an indicator of mitochondrial chelatable Fe2+ in normal hepatocytes with polarized mitochondria as well as in cells undergoing loss of ΔΨm.


Subject(s)
Fluorescent Dyes , Iron Chelating Agents , Mitochondria , Animals , Calcium/metabolism , Cations, Divalent/analysis , Cells, Cultured , Fluorescence , Fluorescent Dyes/chemistry , Iron Chelating Agents/analysis , Mice , Mitochondria/chemistry , Mitochondrial Proteins/chemistry , Oxyquinoline/chemistry , Rhodamine 123 , Thapsigargin/pharmacology
2.
Mol Vis ; 27: 151-160, 2021.
Article in English | MEDLINE | ID: mdl-33907370

ABSTRACT

PURPOSE: Recent reports linking HDAC6 to mitochondrial turnover and neurodegeneration led us to hypothesize that an inhibitor such as Vorinostat (suberoylanilide hydroxamic acid, SAHA) may reduce mitochondrial damage found in retinitis pigmentosa (RP), a progressive neurodegenerative disease of the eye. Here we tested the efficacy of SAHA for its ability to protect photoreceptors in in-vitro and in-situ models of RP. As the stressor, we focused on calcium overload. Calcium is one of the main drivers of cell death, and is associated with rod loss in the rd1 mouse retina, which harbors a mutation in the Pde6b gene similar to that found in human patients suffering from autosomal recessive RP. METHOD: Murine photoreceptor cell line (661W) were exposed to agents that led to calcium stress. Cell survival and redox capacity were measured using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, real-time changes in cellular metabolism were assessed using the Seahorse Biosciences XF24 analyzer, and mitochondrial fission-fusion using imaging. In-situ, neuroprotection was assessed in RPE/retina organ cultures of the rd1 mouse. SAHA effects on cell survival were compared in 661W cells with those of the specific HDAC6 inhibitor tubastatin A, and those on protein acetylation by Western blotting. RESULTS: In stressed 661W cells, SAHA was found to increase cell survival that was associated with improved mitochondrial respiration and reduced mitochondrial fission. The protective effects of SAHA were also observed on photoreceptor cell survival in whole retinal organ explants of the rd1 mouse. Even though tubastatin A was ineffective in increasing cell survival in 661W cells, HDAC6 activity was confirmed in 661W cells after SAHA treatment with protein acetylation specific for HDAC6, defined by an increase in tubulin, but not histone acetylation. CONCLUSIONS: SAHA was found to protect mitochondria from damage, and concomitantly reduced photoreceptor cell death in cell and organ cultures. The lack of activity of tubastatin A suggests that there must be an additional mechanism of action involved in the protective mechanism of SAHA that is responsible for its neuroprotection. Overall, SAHA may be a useful treatment for the prevention of photoreceptor degeneration associated with human RP. The results are discussed in the context of the effects of inhibitors that target different classes and members of the HDAC family and their effects on rod versus cone survival.


Subject(s)
Disease Models, Animal , Histone Deacetylase Inhibitors/therapeutic use , Neuroprotective Agents/therapeutic use , Retinitis Pigmentosa/drug therapy , Vorinostat/therapeutic use , Animals , Blotting, Western , Cell Line , Cell Survival/drug effects , Mice , Mitochondria/drug effects , Mitochondrial Diseases/prevention & control , NADH, NADPH Oxidoreductases/metabolism , Organ Culture Techniques , Photoreceptor Cells, Vertebrate/drug effects , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology
3.
Front Pharmacol ; 11: 577, 2020.
Article in English | MEDLINE | ID: mdl-32457608

ABSTRACT

Gynecologic cancers are among the most lethal cancers found in women, and, advanced stage cancers are still a treatment challenge. Ion channels are known to contribute to cellular homeostasis in all cells and mounting evidence indicates that ion channels could be considered potential therapeutic targets against cancer. Nevertheless, the pharmacologic effect of targeting ion channels in cancer is still understudied. We found that the expression of Kir6.2/SUR2 potassium channel is a potential favorable prognostic factor in gynecologic cancers. Also, pharmacological stimulation of the Kir6.2/SUR2 channel activity with the selective activator molecule minoxidil arrests tumor growth in a xenograft model of ovarian cancer. Investigation on the mechanism linking the Kir6.2/SUR2 to tumor growth revealed that minoxidil alters the metabolic and oxidative state of cancer cells by producing mitochondrial disruption and extensive DNA damage. Consequently, application of minoxidil results in activation of a caspase-3 independent cell death pathway. Our data show that repurposing of FDA approved K+ channel activators may represent a novel, safe adjuvant therapeutic approach to traditional chemotherapy for the treatment of gynecologic cancers.

4.
Nat Biomed Eng ; 4(4): 446-462, 2020 04.
Article in English | MEDLINE | ID: mdl-32284552

ABSTRACT

Environmental factors are the largest contributors to cardiovascular disease. Here we show that cardiac organoids that incorporate an oxygen-diffusion gradient and that are stimulated with the neurotransmitter noradrenaline model the structure of the human heart after myocardial infarction (by mimicking the infarcted, border and remote zones), and recapitulate hallmarks of myocardial infarction (in particular, pathological metabolic shifts, fibrosis and calcium handling) at the transcriptomic, structural and functional levels. We also show that the organoids can model hypoxia-enhanced doxorubicin cardiotoxicity. Human organoids that model diseases with non-genetic pathological factors could help with drug screening and development.


Subject(s)
Drug Evaluation, Preclinical/methods , Heart/drug effects , Models, Cardiovascular , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Organoids/drug effects , Cardiotoxicity/metabolism , Cardiotoxicity/pathology , Drug Development , Humans , Myocardial Infarction/chemically induced , Myocardial Infarction/genetics , Organoids/metabolism , Organoids/pathology , Oxygen/metabolism
5.
Cell Rep ; 28(7): 1879-1893.e7, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31412253

ABSTRACT

Sphingosine 1-phosphate (S1P), a bioactive lysophospholipid generated by sphingosine kinase 1 (SphK1), regulates lymphocyte egress into circulation via S1P receptor 1 (S1PR1) signaling, and it controls the differentiation of regulatory T cells (Tregs) and T helper-17 cells. However, the mechanisms by which receptor-independent SphK1-mediated intracellular S1P levels modulate T cell functionality remains unknown. We show here that SphK1-deficient T cells maintain central memory phenotype and exhibit higher mitochondrial respiration and reduced differentiation to Tregs. Mechanistically, we discovered a direct correlation between SphK1-generated S1P and lipid transcription factor PPARγ (peroxisome proliferator-activated receptor gamma) activity, which in turn regulates lipolysis in T cells. Genetic and pharmacologic inhibition of SphK1 improved metabolic fitness and anti-tumor activity of T cells against murine melanoma. Further, inhibition of SphK1 and PD1 together led to improved control of melanoma. Overall, these data highlight the clinical potential of limiting SphK1/S1P signaling for enhancing anti-tumor-adoptive T cell therapy.


Subject(s)
Cellular Reprogramming , Gene Expression Regulation, Neoplastic , Lysophospholipids/metabolism , Melanoma, Experimental/pathology , PPAR gamma/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Sphingosine/analogs & derivatives , T-Lymphocytes/immunology , Animals , Female , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Phosphorylation , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Sphingosine/metabolism , T-Lymphocytes/metabolism
6.
Sci Rep ; 9(1): 6790, 2019 May 01.
Article in English | MEDLINE | ID: mdl-31043635

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

7.
FASEB J ; 33(7): 8186-8201, 2019 07.
Article in English | MEDLINE | ID: mdl-30951369

ABSTRACT

Statins, widely used to treat hypercholesterolemia, inhibit the 3-hydroxy-3-methylglutaryl-coenzyme A reductase, the rate-limiting enzyme of de novo cholesterol (Chol) synthesis. Statins have been also reported to slow tumor progression. In cancer cells, ATP is generated both by glycolysis and oxidative phosphorylation. Mitochondrial membrane potential (ΔΨ), a readout of mitochondrial metabolism, is sustained by the oxidation of respiratory substrates in the Krebs cycle to generate NADH and flavin adenine dinucleotide, which are further oxidized by the respiratory chain. Here, we studied the short-term effects of statins (3-24 h) on mitochondrial metabolism on cancer cells. Lovastatin (LOV) and simvastatin (SIM) increased ΔΨ in HepG2 and Huh7 human hepatocarcinoma cells and HCC4006 human lung adenocarcinoma cells. Mitochondrial hyperpolarization after LOV and SIM was dose and time dependent. Maximal increase in ΔΨ occurred at 10 µM and 24 h for both statins. The structurally unrelated atorvastatin also hyperpolarized mitochondria in HepG2 cells. Cellular and mitochondrial Chol remained unchanged after SIM. Both LOV and SIM decreased basal respiration, ATP-linked respiration, and ATP production. LOV and SIM did not change the rate of lactic acid production. In summary, statins modulate mitochondrial metabolism in cancer cells independently of the Chol content in cellular membranes without affecting glycolysis.-Christie, C. F., Fang, D., Hunt, E. G., Morris, M. E., Rovini, A., Heslop, K. A., Beeson, G. C., Beeson, C. C., Maldonado, E. N. Statin-dependent modulation of mitochondrial metabolism in cancer cells is independent of cholesterol content.


Subject(s)
Adenocarcinoma of Lung/metabolism , Carcinoma, Hepatocellular/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Liver Neoplasms/metabolism , Lovastatin/pharmacology , Lung Neoplasms/metabolism , Mitochondria, Liver/metabolism , Simvastatin/pharmacology , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/pathology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/pathology
8.
Nat Commun ; 10(1): 1296, 2019 03 21.
Article in English | MEDLINE | ID: mdl-30899002

ABSTRACT

The dysregulation of Fbxo4-cyclin D1 axis occurs at high frequency in esophageal squamous cell carcinoma (ESCC), where it promotes ESCC development and progression. However, defining a therapeutic vulnerability that results from this dysregulation has remained elusive. Here we demonstrate that Rb and mTORC1 contribute to Gln-addiction upon the dysregulation of the Fbxo4-cyclin D1 axis, which leads to the reprogramming of cellular metabolism. This reprogramming is characterized by reduced energy production and increased sensitivity of ESCC cells to combined treatment with CB-839 (glutaminase 1 inhibitor) plus metformin/phenformin. Of additional importance, this combined treatment has potent efficacy in ESCC cells with acquired resistance to CDK4/6 inhibitors in vitro and in xenograft tumors. Our findings reveal a molecular basis for cancer therapy through targeting glutaminolysis and mitochondrial respiration in ESCC with dysregulated Fbxo4-cyclin D1 axis as well as cancers resistant to CDK4/6 inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Gene Expression Regulation, Neoplastic , Glutamine/metabolism , Hypoglycemic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Benzeneacetamides/pharmacology , Cell Line, Tumor , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Drug Resistance, Neoplasm/genetics , Drug Synergism , Energy Metabolism/drug effects , Energy Metabolism/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , F-Box Proteins/genetics , F-Box Proteins/metabolism , Glutaminase/antagonists & inhibitors , Glutaminase/genetics , Glutaminase/metabolism , Glutamine/antagonists & inhibitors , Humans , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Metformin/pharmacology , Mice , Molecular Targeted Therapy , Phenformin/pharmacology , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Signal Transduction , Thiadiazoles/pharmacology , Xenograft Model Antitumor Assays
10.
J Pharmacol Exp Ther ; 369(1): 173-180, 2019 04.
Article in English | MEDLINE | ID: mdl-30709866

ABSTRACT

Acute kidney injury (AKI) is the rapid loss of renal function after an insult, and renal proximal tubule cells (RPTCs) are central to the pathogenesis of AKI. The ß 2-adrenergic receptor (ß 2AR) agonist formoterol accelerates the recovery of renal function in mice after ischemia-reperfusion injury (IRI) with associated rescue of mitochondrial proteins; however, the cell type responsible for this recovery remains unknown. The role of RPTCs in formoterol-induced recovery of renal function was assessed in a proximal tubule-specific knockout of the ß 2AR (γGT-Cre:ADRB2Flox/Flox). These mice and wild-type controls (ADRB2Flox/Flox) were subjected to renal IRI, followed by once-daily dosing of formoterol beginning 24 hours post-IRI and euthanized at 144 hours. Compared with ADRB2Flox/Flox mice, γGT-Cre:ADRB2Flox/Flox mice had decreased renal cortical mRNA expression of the ß 2AR. After IRI, formoterol treatment restored renal function in ADRB2Flox/Flox but not γGT-Cre:ADRB2Flox/Flox mice as measured by serum creatinine, histopathology, and expression of kidney injury marker-1 (KIM-1). Formoterol-treated ADRB2Flox/Flox mice exhibited recovery of mitochondrial proteins and DNA copy number, whereas γGT-Cre:ADRB2Flox/Flox mice treated with formoterol did not. Analysis of mitochondrial morphology by transmission electron microscopy demonstrated that formoterol increased mitochondrial number and density in ADRB2Flox/Flox mice but not in γGT-Cre:ADRB2Flox/Flox mice. These data demonstrate that proximal tubule ß 2AR regulates renal mitochondrial homeostasis. Formoterol accelerates the recovery of renal function after AKI by activating proximal tubule ß 2AR to induce mitochondrial biogenesis and demonstrates the overall requirement of RPTCs in renal recovery.


Subject(s)
Formoterol Fumarate/pharmacology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/physiopathology , Mitochondria/drug effects , Receptors, Adrenergic, beta-2/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Animals , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Mice , Mitochondria/pathology , Recovery of Function/drug effects , Reperfusion Injury/metabolism , Signal Transduction/drug effects
11.
Clin Cancer Res ; 25(3): 1036-1049, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30327305

ABSTRACT

PURPOSE: Adoptive T-cell therapy (ACT) of cancer, which involves the infusion of ex vivo-engineered tumor epitope reactive autologous T cells into the tumor-bearing host, is a potential treatment modality for cancer. However, the durable antitumor response following ACT is hampered either by loss of effector function or survival of the antitumor T cells. Therefore, strategies to improve the persistence and sustain the effector function of the antitumor T cells are of immense importance. Given the role of metabolism in determining the therapeutic efficacy of T cells, we hypothesize that inhibition of PIM kinases, a family of serine/threonine kinase that promote cell-cycle transition, cell growth, and regulate mTORC1 activity, can improve the potency of T cells in controlling tumor. EXPERIMENTAL DESIGN: The role of PIM kinases in T cells was studied either by genetic ablation (PIM1-/-PIM2-/-PIM3-/-) or its pharmacologic inhibition (pan-PIM kinase inhibitor, PimKi). Murine melanoma B16 was established subcutaneously and treated by transferring tumor epitope gp100-reactive T cells along with treatment regimen that involved inhibiting PIM kinases, anti-PD1 or both. RESULTS: With inhibition of PIM kinases, T cells had significant reduction in their uptake of glucose, and upregulated expression of memory-associated genes that inversely correlate with glycolysis. In addition, the expression of CD38, which negatively regulates the metabolic fitness of the T cells, was also reduced in PimKi-treated cells. Importantly, the efficacy of antitumor T-cell therapy was markedly improved by inhibiting PIM kinases in tumor-bearing mice receiving ACT, and further enhanced by adding anti-PD1 antibody to this combination. CONCLUSIONS: This study highlights the potential therapeutic significance of combinatorial strategies where ACT and inhibition of signaling kinase with checkpoint blockade could improve tumor control.


Subject(s)
Biphenyl Compounds/pharmacology , Immunotherapy, Adoptive/methods , Neoplasms, Experimental/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , T-Lymphocytes/immunology , Thiazolidines/pharmacology , Xenograft Model Antitumor Assays/methods , Animals , Antibodies/immunology , Antibodies/pharmacology , Cell Line, Tumor , Humans , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , T-Lymphocytes/metabolism , Treatment Outcome
12.
Cell Rep ; 25(6): 1469-1484.e5, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30404003

ABSTRACT

Patients with mtDNA depletion syndrome 3 (MTDPS3) often die as children from liver failure caused by severe reduction in mtDNA content. The identification of treatments has been impeded by an inability to culture and manipulate MTDPS3 primary hepatocytes. Here we generated DGUOK-deficient hepatocyte-like cells using induced pluripotent stem cells (iPSCs) and used them to identify drugs that could improve mitochondrial ATP production and mitochondrial function. Nicotinamide adenine dinucleotide (NAD) was found to improve mitochondrial function in DGUOK-deficient hepatocyte-like cells by activating the peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α). NAD treatment also improved ATP production in MTDPS3-null rats and in hepatocyte-like cells that were deficient in ribonucleoside-diphosphate reductase subunit M2B (RRM2B), suggesting that it could be broadly effective. Our studies reveal that DGUOK-deficient iPSC-derived hepatocytes recapitulate the pathophysiology of MTDPS3 in culture and can be used to identify therapeutics for mtDNA depletion syndromes.


Subject(s)
DNA, Mitochondrial/genetics , Hepatocytes/metabolism , Induced Pluripotent Stem Cells/metabolism , NAD/metabolism , Adenosine Triphosphate/metabolism , Animals , Base Sequence , Cell Differentiation , Cell Respiration , Female , Glucose/metabolism , Glycolysis , Hepatocytes/cytology , Hepatocytes/ultrastructure , Humans , Induced Pluripotent Stem Cells/cytology , Male , Mitochondria/metabolism , Mitochondria/ultrastructure , Mutation/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Rats , Ribonucleotide Reductases/metabolism , Syndrome
13.
Am J Physiol Renal Physiol ; 315(4): F1119-F1128, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29846105

ABSTRACT

Our laboratory previously reported that agonists of the 5-hydoxytryptamine 1F (5-HT1F) receptor induce renal mitochondrial biogenesis (MB) and that stimulation of the 5-HT1F receptor following ischemia/reperfusion (I/R)-induced acute kidney injury (AKI) accelerated the recovery of renal function in mice. The goal of this study was to examine the contribution of the 5-HT1F receptor in the regulation of renal mitochondrial homeostasis and renal function in naïve and injured mice. Although 5-HT1F receptor knockout (KO) mice were healthy and fertile, and did not exhibit renal dysfunction, renal mitochondrial DNA copy number and mitochondrial fission gene expression increased at 10 wk of age. The 5-HT1F receptor KO mice exhibited greater proximal tubular injury and diminished renal recovery after I/R-induced AKI compared with wild-type mice. These findings were associated with persistent suppression of renal cortical MB and ATP levels after injury. In summary, the 5-HT1F receptor is a component of physiological MB regulation in the kidney, and its absence potentiates renal injury and impedes recovery.


Subject(s)
Acute Kidney Injury/metabolism , Homeostasis/physiology , Mitochondria/metabolism , Receptors, Serotonin/metabolism , Animals , DNA, Mitochondrial/metabolism , Kidney/metabolism , Kidney Cortex/metabolism , Male , Mice, Knockout , Organelle Biogenesis , Receptors, Serotonin/genetics , Reperfusion Injury/metabolism , Receptor, Serotonin, 5-HT1F
14.
Mol Neurobiol ; 55(1): 267-275, 2018 01.
Article in English | MEDLINE | ID: mdl-28889362

ABSTRACT

Myelin basic protein (MBP) is a major target of T cells in lesions of multiple sclerosis (MS) patients and its animal model, experimental autoimmune encephalomyelitis (EAE). Interactions between the major histocompatibility complex II containing antigenic peptides and the T cell receptor activate CD4+ T cells that perpetuate EAE and MS. Previously reported data has shown that treating with an altered peptide ligand (APL) in which the normal antigenic peptide sequence of MBP has been slightly changed at T cell contact positions is helpful in reducing disease in both rodents and humans. The use of natural peptides, which are susceptible to protease degradation, requires high concentrations that can create hypersensitivity reactions. Our hypothesis is that APL containing aza substitutions, CH(R)-N- > N(R)N, could lead to improved protease resistance, reduced clinical disease scores, and a shift in T cell profile. In this study, several aza-APLs and control peptides were synthesized and screened for the best aza-APL candidate (3aza-APL) based on dissociation half time from major histocompatibility complex (MHC) class II, induction of IL-2 response, and resistance to degradation by proteases. The efficacy was then tested in vivo. Results indicate that 3aza-APL is superior to currently available APLs in terms of protease resistance and disease suppression in EAE mice. The 3aza-APL induced anti-inflammatory immune responses by altering key transcription factors and cytokine genes which regulate T cell subpopulations. These data suggest that the novel 3aza-APL has increased protease resistance property and is effective in reducing clinical and physiological signs of disease in EAE animals.


Subject(s)
Aza Compounds/administration & dosage , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Myelin Basic Protein/administration & dosage , Animals , Cell Line , Encephalomyelitis, Autoimmune, Experimental/pathology , Male , Mice , Treatment Outcome
15.
Cell Metab ; 27(1): 85-100.e8, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29129787

ABSTRACT

Heightened effector function and prolonged persistence, the key attributes of Th1 and Th17 cells, respectively, are key features of potent anti-tumor T cells. Here, we established ex vivo culture conditions to generate hybrid Th1/17 cells, which persisted long-term in vivo while maintaining their effector function. Using transcriptomics and metabolic profiling approaches, we showed that the enhanced anti-tumor property of Th1/17 cells was dependent on the increased NAD+-dependent activity of the histone deacetylase Sirt1. Pharmacological or genetic inhibition of Sirt1 activity impaired the anti-tumor potential of Th1/17 cells. Importantly, T cells with reduced surface expression of the NADase CD38 exhibited intrinsically higher NAD+, enhanced oxidative phosphorylation, higher glutaminolysis, and altered mitochondrial dynamics that vastly improved tumor control. Lastly, blocking CD38 expression improved tumor control even when using Th0 anti-tumor T cells. Thus, strategies targeting the CD38-NAD+ axis could increase the efficacy of anti-tumor adoptive T cell therapy.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Immunotherapy , NAD/metabolism , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Forkhead Box Protein O1/metabolism , Glutamine/metabolism , Mice, Inbred C57BL , Neoplasms/metabolism , Sirtuin 1/metabolism , Th1 Cells/immunology , Th17 Cells/immunology
16.
Am J Physiol Renal Physiol ; 314(2): F260-F268, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29046298

ABSTRACT

Our laboratory recently made the novel observation that 5-hydroxytryptamine 1F (5-HT1F) receptor activation induces mitochondrial biogenesis (MB), the production of new, functional mitochondria, in vitro and in vivo. We sought to determine the mechanism linking the 5-HT1F receptor to MB in renal proximal tubule cells. Using LY344864 , a selective 5-HT1F receptor agonist, we determined that the 5-HT1F receptor is coupled to Gαi/o and induces MB through Gßγ-dependent activation of Akt, endothelial nitric oxide synthase (eNOS), cyclic guanosine-monophosphate (cGMP), protein kinase G (PKG), and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α). We also report that the 5-HT1F receptor signals through a second, Gßγ-dependent pathway that is linked by Akt phosphorylation of Raf. In contrast to the activated Akt pathway, Raf phosphorylation reduced extracellular signal regulated kinases (ERK1/2) and foxhead box O3a (FOXO3a) phosphorylation, suppressing an inhibitory MB pathway. These results demonstrate that the 5-HT1F receptor regulates MB through Gßγ-dependent dual mechanisms that activate a stimulatory MB pathway, Akt/eNOS/cGMP/PKG/PGC-1α, while simultaneously repressing an inhibitory MB pathway, Raf/MEK/ERK/FOXO3a. Novel mechanisms of MB provide the foundation for new chemicals that induce MB to treat acute and chronic organ injuries.


Subject(s)
Kidney Tubules, Proximal/metabolism , Mitochondria/metabolism , Organelle Biogenesis , Receptors, Serotonin/metabolism , Animals , Carbazoles/pharmacology , Cells, Cultured , Female , Fluorobenzenes/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Tubules, Proximal/drug effects , Mitochondria/drug effects , Phosphorylation , Rabbits , Receptors, Serotonin/drug effects , Second Messenger Systems , Serotonin 5-HT1 Receptor Agonists/pharmacology , Receptor, Serotonin, 5-HT1F
17.
J Mol Cell Cardiol ; 114: 309-319, 2018 01.
Article in English | MEDLINE | ID: mdl-29224834

ABSTRACT

RATIONALE: Recent evidence indicates that histone deacetylase enzymes (HDACs) contribute to ischemia reperfusion (I/R) injury, and pan-HDAC inhibitors have been shown to be cardioprotective when administered either before an ischemic insult or during reperfusion. We have shown previously that selective inhibition of class I HDACs provides superior cardioprotection when compared to pan-HDAC inhibition in a pretreatment model, but selective class I HDAC inhibition has not been tested during reperfusion, and specific targets of class I HDACs in I/R injury have not been identified. OBJECTIVE: We hypothesized that selective inhibition of class I HDACs with the drug MS-275 (entinostat) during reperfusion would improve recovery from I/R injury in the first hour of reperfusion. METHODS AND RESULTS: Hearts from male Sprague-Dawley rats were subjected to ex vivo I/R injury±MS-275 class I HDAC inhibition during reperfusion alone. MS-275 significantly attenuated I/R injury, as indicated by improved LV function and tissue viability at the end of reperfusion. Unexpectedly, we observed that HDAC1 is present in the mitochondria of cardiac myocytes, but not fibroblasts or endothelial cells. We then designed mitochondria-restricted and mitochondria-excluded HDAC inhibitors, and tested both in our ex vivo I/R model. The selective inhibition of mitochondrial HDAC1 attenuated I/R injury to the same extent as MS-275, whereas the mitochondrial-excluded inhibitor did not. Further assays demonstrated that these effects are attributable to a decrease in SDHA activity and subsequent metabolic ROS production in reperfusion. CONCLUSIONS: We demonstrate for the first time that HDAC1 is present within the mitochondria of cardiac myocytes, and mitochondrial HDAC1 contributes significantly to I/R injury within the first hour of reperfusion.


Subject(s)
Mitochondria/enzymology , Myocardial Reperfusion Injury/enzymology , Myocytes, Cardiac/enzymology , Animals , Cell Survival/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Male , Mitochondria/drug effects , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/enzymology , Myocardium/pathology , Myocytes, Cardiac/pathology , Oxygen Consumption/drug effects , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Succinate Dehydrogenase/metabolism , Ventricular Function/drug effects
18.
Sci Rep ; 7(1): 10578, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28874749

ABSTRACT

Mitochondrial dysfunction is associated with numerous acute and chronic degenerative diseases. The beta-2 adrenergic receptor (ß2AR) agonist formoterol induces mitochondrial biogenesis (MB), but other ß2AR agonists, such as clenbuterol, do not. We sought to identify the MB signaling pathway of formoterol and the differences in signaling between these two ligands that result in the differential induction of MB. While formoterol and clenbuterol increased cAMP, only formoterol increased the phosphorylation of Akt and its downstream target eNOS. The increase in Akt phosphorylation was Gßγ- and PI3K-dependent, and the increase in eNOS phosphorylation was Gßγ- and Akt-dependent. Only formoterol increased cGMP. Formoterol induced MB as measured by increases in uncoupled cellular respiration and PGC-1α and NDUFS1 mRNA expression and was blocked by inhibitors of Gßγ, Akt, NOS, and soluble guanylate cyclase. To identify distinct receptor-ligand interactions leading to these differences in signaling, we docked formoterol and clenbuterol to six structures of the ß2AR. Compared to clenbuterol, the methoxyphenyl group of formoterol interacted more frequently with V114 and F193, while its formamide group interacted more frequently with C191. These data indicate that the unique structural features of formoterol allow it to interact with the ß2AR to activate the Gßγ-Akt-eNOS-sGC pathway to induce MB.


Subject(s)
Clenbuterol/chemistry , Clenbuterol/pharmacology , Formoterol Fumarate/chemistry , Formoterol Fumarate/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Organelle Biogenesis , Animals , Cell Respiration/drug effects , Cyclic GMP/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Kidney Tubules, Proximal/cytology , Models, Molecular , Molecular Conformation , Oxygen Consumption , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rabbits
19.
Cancer Immunol Res ; 5(8): 666-675, 2017 08.
Article in English | MEDLINE | ID: mdl-28642246

ABSTRACT

The endoplasmic reticulum (ER) is an energy-sensing organelle with intimate ties to programming cell activation and metabolic fate. T-cell receptor (TCR) activation represents a form of acute cell stress and induces mobilization of ER Ca2+ stores. The role of the ER in programming T-cell activation and metabolic fate remains largely undefined. Gp96 is an ER protein with functions as a molecular chaperone and Ca2+ buffering protein. We hypothesized that the ER stress response may be important for CD4+ T-cell activation and that gp96 may be integral to this process. To test our hypothesis, we utilized genetic deletion of the gp96 gene Hsp90b1 in a CD4+ T cell-specific manner. We show that gp96-deficient CD4+ T cells cannot undergo activation-induced glycolysis due to defective Ca2+ mobilization upon TCR engagement. We found that activating naïve CD4+ T cells while inhibiting ER Ca2+ exchange, through pharmacological blockade of the ER Ca2+ channel inositol trisphosphate receptor (IP3R), led to a reduction in cytosolic Ca2+ content and generated a pool of CD62Lhigh/CD44low CD4+ T cells compared with wild-type (WT) matched controls. In vivo IP3R-inhibited CD4+ T cells exhibited elevated tumor control above WT T cells. Together, these data show that ER-modulated cytosolic Ca2+ plays a role in defining CD4+ T-cell phenotype and function. Factors associated with the ER stress response are suitable targets for T cell-based immunotherapies. Cancer Immunol Res; 5(8); 666-75. ©2017 AACR.


Subject(s)
Endoplasmic Reticulum Stress/immunology , Membrane Glycoproteins/immunology , Neoplasms/immunology , Receptors, Antigen, T-Cell/immunology , CD4-Positive T-Lymphocytes/immunology , Calcium/metabolism , Endoplasmic Reticulum Stress/genetics , Glycolysis , Humans , Hyaluronan Receptors/immunology , Inositol 1,4,5-Trisphosphate Receptors/immunology , L-Selectin/immunology , Lymphocyte Activation/immunology , Membrane Glycoproteins/genetics , Neoplasms/pathology , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics
20.
J Biol Chem ; 292(25): 10465-10489, 2017 06 23.
Article in English | MEDLINE | ID: mdl-28389562

ABSTRACT

The appearance of myofibroblasts is generally thought to be the underlying cause of the fibrotic changes that underlie idiopathic pulmonary fibrosis. However, the cellular/molecular mechanisms that account for the fibroblast-myofibroblast differentiation/activation in idiopathic pulmonary fibrosis remain poorly understood. We investigated the functional role of hyaluronan receptor CD44V6 (CD44 containing variable exon 6 (v6)) for differentiation of lung fibroblast to myofibroblast phenotype. Increased hyaluronan synthesis and CD44 expression have been detected in numerous fibrotic organs. Previously, we found that the TGFß1/CD44V6 pathway is important in lung myofibroblast collagen-1 and α-smooth-muscle actin synthesis. Because increased EGR1 (early growth response-1) expression has been shown to appear very early and nearly coincident with the expression of CD44V6 found after TGFß1 treatment, we investigated the mechanism(s) of regulation of CD44V6 expression in lung fibroblasts by TGFß1. TGFß1-mediated CD44V6 up-regulation was initiated through EGR1 via ERK-regulated transcriptional activation. We showed that TGFß1-induced CD44V6 expression is through EGR1-mediated AP-1 (activator protein-1) activity and that the EGR1- and AP-1-binding sites in the CD44v6 promoter account for its responsiveness to TGFß1 in lung fibroblasts. We also identified a positive-feedback loop in which ERK/EGR1 signaling promotes CD44V6 splicing and found that CD44V6 then sustains ERK signaling, which is important for AP-1 activity in lung fibroblasts. Furthermore, we identified that HAS2-produced hyaluronan is required for CD44V6 and TGFßRI co-localization and subsequent CD44V6/ERK1/EGR1 signaling. These results demonstrate a novel positive-feedback loop that links the myofibroblast phenotype to TGFß1-stimulated CD44V6/ERK/EGR1 signaling.


Subject(s)
Early Growth Response Protein 1/metabolism , Hyaluronan Receptors/biosynthesis , Lung/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 3/metabolism , Myofibroblasts/metabolism , Pulmonary Fibrosis/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Gene Expression Regulation , Glucuronosyltransferase/metabolism , Hyaluronan Synthases , Hyaluronic Acid/biosynthesis , Lung/pathology , Mice , Myofibroblasts/pathology , Pulmonary Fibrosis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...