Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cells ; 10(6)2021 05 21.
Article in English | MEDLINE | ID: mdl-34064003

ABSTRACT

Ceramide and diacylglycerol (DAG) are bioactive lipids and mediate many cellular signaling pathways. Sphingomyelin synthase (SMS) is the single metabolic link between the two, while SMS2 is the only SMS form located at the plasma membrane. SMS2 functions were investigated in HepG2 cell lines stably expressing SMS2. SMS2 overexpression did not alter sphingomyelin (SM), phosphatidylcholine (PC), or ceramide levels. DAG content increased by approx. 40% and led to downregulation of DAG-dependent protein kinase C (PKC). SMS2 overexpression also induced senescence, characterized by positivity for ß-galactosidase activity and heterochromatin foci. HepG2-SMS2 cells exhibited protruded mitochondria and suppressed mitochondrial respiration rates. ATP production and the abundance of Complex V were substantially lower in HepG2-SMS2 cells as compared to controls. SMS2 overexpression was associated with inflammasome activation based on increases in IL-1ß and nlpr3 mRNA levels. HepG2-SMS2 cells exhibited lipid droplet accumulation, constitutive activation of AMPK based on elevated 172Thr phosphorylation, increased AMPK abundance, and insensitivity to insulin suppression of AMPK. Thus, our results show that SMS2 regulates DAG homeostasis and signaling in hepatocytes and also provide proof of principle for the concept that offset in bioactive lipids' production at the plasma membrane can drive the senescence program in association with steatosis and, seemingly, by cell-autonomous mechanisms.


Subject(s)
Cell Membrane/metabolism , Ceramides/metabolism , Diglycerides/metabolism , Transferases (Other Substituted Phosphate Groups)/metabolism , Cellular Senescence , Fatty Liver/metabolism , Hep G2 Cells , Humans
2.
Biochim Biophys Acta Mol Basis Dis ; 1866(10): 165883, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32592935

ABSTRACT

Juvenile neuronal ceroid lipofuscinosis (JNCL, aka. juvenile Batten disease or CLN3 disease) is a lysosomal storage disease characterized by progressive blindness, seizures, cognitive and motor failures, and premature death. JNCL is caused by mutations in the Ceroid Lipofuscinosis, Neuronal 3 (CLN3) gene, whose function is unclear. Although traditionally considered a neurodegenerative disease, CLN3 disease displays eye-specific effects: Vision loss not only is often one of the earliest symptoms of JNCL, but also has been reported in non-syndromic CLN3 disease. Here we described the roles of CLN3 protein in maintaining healthy retinal pigment epithelium (RPE) and normal vision. Using electroretinogram, fundoscopy and microscopy, we showed impaired visual function, retinal autofluorescent lesions, and RPE disintegration and metaplasia/hyperplasia in a Cln3 ~ 1 kb-deletion mouse model [1] on C57BL/6J background. Utilizing a combination of biochemical analyses, RNA-Seq, Seahorse XF bioenergetic analysis, and Stable Isotope Resolved Metabolomics (SIRM), we further demonstrated that loss of CLN3 increased autophagic flux, suppressed mTORC1 and Akt activities, enhanced AMPK activity, and up-regulated gene expression of the autophagy-lysosomal system in RPE-1 cells, suggesting autophagy induction. This CLN3 deficiency induced autophagy induction coincided with decreased mitochondrial oxygen consumption, glycolysis, the tricarboxylic acid (TCA) cycle, and ATP production. We also reported for the first time that loss of CLN3 led to glycogen accumulation despite of impaired glycogen synthesis. Our comprehensive analyses shed light on how loss of CLN3 affect autophagy and metabolism. This work suggests possible links among metabolic impairment, autophagy induction and lysosomal storage, as well as between RPE atrophy/degeneration and vision loss in JNCL.


Subject(s)
Blindness/genetics , Membrane Glycoproteins/deficiency , Neuronal Ceroid-Lipofuscinoses/genetics , Retinal Pigment Epithelium/pathology , Animals , Atrophy/genetics , Atrophy/pathology , Autophagy , Blindness/pathology , Cell Line , Disease Models, Animal , Gene Knock-In Techniques , Gene Knockdown Techniques , Glycogen/metabolism , Humans , Lysosomes/pathology , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Microscopy, Electron , Molecular Chaperones/genetics , Mutation , Neuronal Ceroid-Lipofuscinoses/complications , Neuronal Ceroid-Lipofuscinoses/pathology , RNA, Small Interfering/metabolism , Retinal Pigment Epithelium/ultrastructure
3.
J Biol Chem ; 294(18): 7516-7527, 2019 05 03.
Article in English | MEDLINE | ID: mdl-30885944

ABSTRACT

It is generally accepted that alterations in metabolism are critical for the metastatic process; however, the mechanisms by which these metabolic changes are controlled by the major drivers of the metastatic process remain elusive. Here, we found that S100 calcium-binding protein A4 (S100A4), a major metastasis-promoting protein, confers metabolic plasticity to drive tumor invasion and metastasis of non-small cell lung cancer cells. Investigating how S100A4 regulates metabolism, we found that S100A4 depletion decreases oxygen consumption rates, mitochondrial activity, and ATP production and also shifts cell metabolism to higher glycolytic activity. We further identified that the 49-kDa mitochondrial complex I subunit NADH dehydrogenase (ubiquinone) Fe-S protein 2 (NDUFS2) is regulated in an S100A4-dependent manner and that S100A4 and NDUFS2 exhibit co-occurrence at significant levels in various cancer types as determined by database-driven analysis of genomes in clinical samples using cBioPortal for Cancer Genomics. Importantly, we noted that S100A4 or NDUFS2 silencing inhibits mitochondrial complex I activity, reduces cellular ATP level, decreases invasive capacity in three-dimensional growth, and dramatically decreases metastasis rates as well as tumor growth in vivo Finally, we provide evidence that cells depleted in S100A4 or NDUFS2 shift their metabolism toward glycolysis by up-regulating hexokinase expression and that suppressing S100A4 signaling sensitizes lung cancer cells to glycolysis inhibition. Our findings uncover a novel S100A4 function and highlight its importance in controlling NDUFS2 expression to regulate the plasticity of mitochondrial metabolism and thereby promote the invasive and metastatic capacity in lung cancer.


Subject(s)
Lung Neoplasms/metabolism , Lung Neoplasms/pathology , NADH Dehydrogenase/metabolism , Neoplasm Invasiveness , S100 Calcium-Binding Protein A4/metabolism , Up-Regulation , Adenosine Triphosphate/biosynthesis , Cell Line, Tumor , Gene Silencing , Glycolysis , Humans , NADH Dehydrogenase/genetics , Neoplasm Metastasis , Signal Transduction
4.
Transplantation ; 103(4): 705-715, 2019 04.
Article in English | MEDLINE | ID: mdl-30451741

ABSTRACT

BACKGROUND: Experimental and preclinical evidence suggest that adoptive transfer of regulatory T (Treg) cells could be an appropriate therapeutic strategy to induce tolerance and improve graft survival in transplanted patients. The University of Kentucky Transplant Service Line is developing a novel phase I/II clinical trial with ex vivo expanded autologous Treg cells as an adoptive cellular therapy in renal transplant recipients who are using everolimus (EVR)-based immunosuppressive regimen. METHODS: The aim of this study was to determine the mechanisms of action and efficacy of EVR for the development of functionally competent Treg cell-based adoptive immunotherapy in transplantation to integrate a common EVR-based regimen in vivo (in the patient) and ex vivo (in the expansion of autologous Treg cells). CD25 Treg cells were selected from leukapheresis product with a GMP-compliant cell separation system and placed in 5-day (short) or 21-day (long) culture with EVR or rapamycin (RAPA). Multi-parametric flow cytometry analyses were used to monitor the expansion rates, phenotype, autophagic flux, and suppressor function of the cells. phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin signaling pathway profiles of treated cells were analyzed by Western blot and cell bioenergetic parameters by extracellular flux analysis. RESULTS: EVR-treated cells showed temporary slower growth, lower metabolic rates, and reduced phosphorylation of protein kinase B compared with RAPA-treated cells. In spite of these differences, the expansion rates, phenotype, and suppressor function of long-term Treg cells in culture with EVR were similar to those with RAPA. CONCLUSIONS: Our results support the feasibility of EVR to expand functionally competent Treg cells for their clinical use.


Subject(s)
Everolimus/pharmacology , Immunosuppressive Agents/pharmacology , Organ Transplantation , T-Lymphocytes, Regulatory/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Cells, Cultured , Energy Metabolism , Flow Cytometry , Humans , Immunotherapy, Adoptive , Membrane Potential, Mitochondrial , Signal Transduction/physiology , Sirolimus/pharmacology , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases/physiology
5.
Transpl Int ; 30(10): 1061-1074, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28543637

ABSTRACT

The PI3K/mTOR signaling cascade is fundamental in T-cell activation and fate decisions. We showed the distinct regulation of PI3K/mTOR in regulatory and effector T-cells and proposed the potential therapeutic benefit of targeting this pathway to control the balance between effector and regulatory T-cell activities. Substantial adverse effects in long-term clinical usage of rapamycin suggest the use of alternative treatments in restraining effector T-cell function in transplant patients. We hypothesize that dual PI3K/mTOR inhibitors may represent an immunosuppressant alternative. Here we show that dual PI3K/mTOR PI-103 and PKI-587 inhibitors interfered IL-2-dependent responses in T-cells. However, in contrast to the inhibitory effects in non-Treg T-cell proliferation and effector functions, dual inhibitors increased the differentiation, preferential expansion, and suppressor activity of iTregs. Rapamycin, PI-103, and PKI-587 targeted different signaling events and induced different metabolic patterns in primary T-cells. Similar to rapamycin, in vivo administration of PI-103 and PKI-587 controlled effectively the immunological response against allogeneic skin graft. These results characterize specific regulatory mechanisms of dual PI3K/mTOR inhibitors in T-cells and support their potential as a novel therapeutic option in transplantation.


Subject(s)
Furans/pharmacology , Morpholines/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , T-Lymphocytes/drug effects , Transplantation Immunology , Triazines/pharmacology , Animals , Drug Evaluation, Preclinical , Humans , Interleukin-2/metabolism , Mice , Phosphoinositide-3 Kinase Inhibitors , Sirolimus , TOR Serine-Threonine Kinases/antagonists & inhibitors
6.
Exp Cell Res ; 354(2): 112-121, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28342898

ABSTRACT

Thermotherapy, as a method of treating cancer, has recently attracted considerable attention from basic and clinical investigators. A number of studies and clinical trials have shown that thermotherapy can be successfully used as a therapeutic approach for various cancers. However, the effects of temperature on cancer bioenergetics have not been studied in detail with a real time, microplate based, label-free detection approach. This study investigates how changes in temperature affect the bioenergetics characteristics (mitochondrial function and glycolysis) of three colorectal cancer (CRC) cell lines utilizing the Seahorse XF96 technology. Experiments were performed at 32°C, 37°C and 42°C using assay medium conditions and equipment settings adjusted to produce equal oxygen and pH levels ubiquitously at the beginning of all experiments. The results suggest that temperature significantly changes multiple components of glycolytic and mitochondrial function of all cell lines tested. Under hypothermia conditions (32°C), the extracellular acidification rates (ECAR) of CRC cells were significantly lower compared to the same basal ECAR levels measured at 37°C. Mitochondrial stress test for SW480 cells at 37°C vs 42°C demonstrated increased proton leak while all other OCR components remained unchanged (similar results were detected also for the patient-derived xenograft cells Pt.93). Interestingly, the FCCP dose response at 37°C vs 42°C show significant shifts in profiles, suggesting that single dose FCCP experiments might not be sufficient to characterize the mitochondrial metabolic potential when comparing groups, conditions or treatments. These findings provide valuable insights for the metabolic and bioenergetic changes of CRC cells under hypo- and hyperthermia conditions that could potentially lead to development of better targeted and personalized strategies for patients undergoing combined thermotherapy with chemotherapy.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Glycolysis , Mitochondria/metabolism , Temperature , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cell Line, Tumor , Cell Respiration/drug effects , Energy Metabolism/drug effects , Glycolysis/drug effects , Humans , Hypothermia, Induced , Mitochondria/drug effects , Oxidative Phosphorylation/drug effects , Oxygen/metabolism , Phenotype , Stress, Physiological/drug effects
7.
Cell Death Dis ; 8(2): e2593, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28151470

ABSTRACT

Obesity has been associated with increased incidence and mortality of a wide variety of human cancers including colorectal cancer. However, the molecular mechanism by which adipocytes regulate the metabolism of colon cancer cells remains elusive. In this study, we showed that adipocytes isolated from adipose tissues of colon cancer patients have an important role in modulating cellular metabolism to support tumor growth and survival. Abundant adipocytes were found in close association with invasive tumor cells in colon cancer patients. Co-culture of adipocytes with colon cancer cells led to a transfer of free fatty acids that released from the adipocytes to the cancer cells. Uptake of fatty acids allowed the cancer cells to survive nutrient deprivation conditions by upregulating mitochondrial fatty acid ß-oxidation. Mechanistically, co-culture of adipocytes or treating cells with fatty acids induced autophagy in colon cancer cells as a result of AMPK activation. Inhibition of autophagy attenuated the ability of cancer cells to utilize fatty acids and blocked the growth-promoting effect of adipocytes. In addition, we found that adipocytes stimulated the expression of genes associated with cancer stem cells and downregulated genes associated with intestinal epithelial cell differentiation in primary colon cancer cells and mouse tumor organoids. Importantly, the presence of adipocytes promoted the growth of xenograft tumors in vivo. Taken together, our results show that adipocytes in the tumor microenvironment serve as an energy provider and a metabolic regulator to promote the growth and survival of colon cancer cells.


Subject(s)
Adipocytes/metabolism , Autophagy/physiology , Colonic Neoplasms/metabolism , Fatty Acids/metabolism , Mitochondria/metabolism , AMP-Activated Protein Kinases/metabolism , Adipocytes/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Autophagy/genetics , Cell Differentiation/genetics , Cell Line , Cell Line, Tumor , Coculture Techniques/methods , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Down-Regulation/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression/genetics , Humans , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Mice , Mitochondria/genetics , Mitochondria/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oxidation-Reduction , Tumor Microenvironment/genetics
8.
Dig Liver Dis ; 49(6): 697-704, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28179093

ABSTRACT

Treatment of advanced hepatocellular carcinoma (HCC) remains a challenge due to the high tumor heterogeneity. In the present study, we aim to evaluate the impact of the ß-catenin inhibitor, FH535, alone or in combination with the Ras/Raf/MAPK inhibitor Sorafenib, on the bioenergetics profiles of the HCC cell lines Huh7 and PLC/PRF/5. Single low-dose treatments with FH535 or Sorafenib promoted different effects on mitochondrial respiration and glycolysis in a cell type specific manner. However, the combination of these drugs significantly reduced both mitochondrial respiration and glycolytic rates regardless of the HCC cells. The significant changes in mitochondrial respiration observed in cells treated with the Sorafenib-FH535 combination may correspond to differential targeting of ETC complexes and changes in substrate utilization mediated by each drug. Moreover, the bioenergetics changes and the loss of mitochondrial membrane potential that were evidenced by treatment of HCC cells with the combination of FH535 and Sorafenib, preceded the induction of cell apoptosis. Overall, our results demonstrated that Sorafenib-FH535 drug combination induce the disruption of the bioenergetics of HCC by the simultaneous targeting of mitochondrial respiration and glycolytic flux that leads the synergistic effect on inhibition of cell proliferation. These findings support the therapeutic potential of combinatory FH535-Sorafenib treatment of the HCC heterogeneity by the simultaneous targeting of different molecular pathways.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Mitochondria/drug effects , Niacinamide/analogs & derivatives , Phenylurea Compounds/administration & dosage , Sulfonamides/administration & dosage , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Energy Metabolism/drug effects , Humans , Liver Neoplasms/pathology , Niacinamide/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Signal Transduction/drug effects , Sorafenib , beta Catenin/antagonists & inhibitors
9.
Cell Death Discov ; 3: 16103, 2017.
Article in English | MEDLINE | ID: mdl-28179998

ABSTRACT

Increased glucose metabolism is considered as one of the most important metabolic alterations adapted by cancer cells in order to generate energy as well as high levels of glycolytic intermediates to support rapid proliferation. PH domain leucine-rich repeat protein phosphatase (PHLPP) belongs to a novel family of Ser/Thr protein phosphatases that function as tumor suppressors in various types of human cancer. Here we determined the role of PHLPP in regulating glucose metabolism in colon cancer cells. Knockdown of PHLPP increased the rate of glucose consumption and lactate production, whereas overexpression of PHLPP had the opposite effect. Bioenergetic analysis using Seahorse Extracelluar Flux Analyzer revealed that silencing PHLPP expression induced a glycolytic shift in colon cancer cells. Mechanistically, we found that PHLPP formed a complex with Akt and hexokinase 2 (HK2) in the mitochondrial fraction of colon cancer cells and knockdown of PHLPP enhanced Akt-mediated phosphorylation and mitochondrial localization of HK2. Depletion of HK2 expression or treating cells with Akt and HK2 inhibitors reversed PHLPP loss-induced increase in glycolysis. Furthermore, PHLPP knockdown cells became addicted to glucose as a major energy source in that glucose starvation significantly decreased cancer cell survival. As HK2 is the key enzyme that determines the direction and magnitude of glucose flux, our study identified PHLPP as a novel regulator of glucose metabolism by controlling HK2 activity in colon cancer cells.

10.
Int J Pharm ; 511(2): 1012-21, 2016 Sep 25.
Article in English | MEDLINE | ID: mdl-27492022

ABSTRACT

Mitochondria are considered to be the "power plants" of the cell, but can also initiate and execute cell death, stimulated by oxidative stress (OS). OS induced mitochondrial dysfunction is characterized by a loss in oxygen consumption and reduced ATP production. Curcumin, as a potential therapeutic, has been explored as a candidate for mitochondrial OS suppression, but rapid metabolism and aqueous insolubility has prevented it from being effective. Further, efficient delivery of curcumin via the incorporation into nanocarriers has again been limited due to low drug loading capacities and/or significant burst release, resulting in acute cytotoxicity. Hence, to increase the therapeutic potential and reduce the toxic effects of curcumin, curcumin conjugated poly(ß-amino ester) nanogels (CNGs) were synthesized using Michael addition chemistry. This approach provided easy control over the nanogel size, with CNGs showing a uniform release of active curcumin over 48h with no burst release. This controlled release system significantly increased the safety limit for curcumin, with a ten fold increase in the cytotoxic threshold, as compared to free curcumin. Further, real-time mitochondrial response analysis with the Seahorse XF96 showed effective and prolonged suppression of H2O2 induced mitochondrial oxidative stress upon pre-treating endothelial cells with CNGs and this potential of nanogels was studied at different pre-treatment times prior to H2O2 exposure.


Subject(s)
Curcumin/administration & dosage , Cytoprotection/drug effects , Mitochondria/drug effects , Oxidative Stress/drug effects , Polyethylene Glycols/administration & dosage , Polyethyleneimine/administration & dosage , Polymers/administration & dosage , Cell Survival/drug effects , Cell Survival/physiology , Curcumin/chemistry , Cytoprotection/physiology , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Dose-Response Relationship, Drug , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mitochondria/metabolism , Nanogels , Oxidative Stress/physiology , Polyethylene Glycols/chemistry , Polyethyleneimine/chemistry , Polymers/chemistry
11.
Biomaterials ; 105: 127-135, 2016 10.
Article in English | MEDLINE | ID: mdl-27521615

ABSTRACT

To increase the efficacy of radiation, iron oxide nanoparticles can be utilized for their ability to produce reactive oxygen species (ROS). Radiation therapy promotes leakage of electrons from the electron transport chain and leads to an increase in mitochondrial production of the superoxide anion which is converted to hydrogen peroxide by superoxide dismutase. Iron oxide nanoparticles can then catalyze the reaction from hydrogen peroxide to the highly reactive hydroxyl radical. Therefore, the overall aim of this project was to utilize iron oxide nanoparticles conjugated to a cell penetrating peptide, TAT, to escape lysosomal encapsulation after internalization by cancer cells and catalyze hydroxyl radical formation. It was determined that TAT functionalized iron oxide nanoparticles and uncoated iron oxide nanoparticles resulted in permeabilization of the lysosomal membranes. Additionally, mitochondrial integrity was compromised when A549 cells were treated with both TAT-functionalized nanoparticles and radiation. Pre-treatment with TAT-functionalized nanoparticles also significantly increased the ROS generation associated with radiation. A long term viability study showed that TAT-functionalized nanoparticles combined with radiation resulted in a synergistic combination treatment. This is likely due to the TAT-functionalized nanoparticles sensitizing the cells to subsequent radiation therapy, because the nanoparticles alone did not result in significant toxicities.


Subject(s)
Ferric Compounds/administration & dosage , Ferrosoferric Oxide/administration & dosage , Gene Products, tat/pharmacokinetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/radiotherapy , Radiation-Sensitizing Agents/administration & dosage , Radiotherapy, Conformal/methods , A549 Cells , Cell Survival/radiation effects , Gene Products, tat/administration & dosage , Humans , Molecular Targeted Therapy/methods , Nanocapsules/administration & dosage , Nanocapsules/chemistry , Neoplasms, Experimental/pathology , Radiation Tolerance , Reactive Oxygen Species/metabolism , Treatment Outcome
12.
VAD J ; 2(1)2016.
Article in English | MEDLINE | ID: mdl-28042604

ABSTRACT

This manuscript describes a protocol at the University of Kentucky that allows a translational research team to collect human myocardium that can be used for biological research. We have gained a great deal of practical experience since we started this protocol in 2008, and we hope that other groups might be able to learn from our endeavors. To date, we have procured ~4000 samples from ~230 patients. The tissue that we collect comes from organ donors and from patients who are receiving a heart transplant or a ventricular assist device because they have heart failure. We begin our manuscript by describing the importance of human samples in cardiac research. Subsequently, we describe the process for obtaining consent from patients, the cost of running the protocol, and some of the issues and practical difficulties that we have encountered. We conclude with some suggestions for other researchers who may be considering starting a similar protocol.

13.
Oncotarget ; 6(22): 18891-904, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-25970773

ABSTRACT

Fatty acid synthase (FASN), a lipogenic enzyme, is upregulated in colorectal cancer (CRC). Increased de novo lipid synthesis is thought to be a metabolic adaptation of cancer cells that promotes survival and metastasis; however, the mechanisms for this phenomenon are not fully understood. We show that FASN plays a role in regulation of energy homeostasis by enhancing cellular respiration in CRC. We demonstrate that endogenously synthesized lipids fuel fatty acid oxidation, particularly during metabolic stress, and maintain energy homeostasis. Increased FASN expression is associated with a decrease in activation of energy-sensing pathways and accumulation of lipid droplets in CRC cells and orthotopic CRCs. Immunohistochemical evaluation demonstrated increased expression of FASN and p62, a marker of autophagy inhibition, in primary CRCs and liver metastases compared to matched normal colonic mucosa. Our findings indicate that overexpression of FASN plays a crucial role in maintaining energy homeostasis in CRC via increased oxidation of endogenously synthesized lipids. Importantly, activation of fatty acid oxidation and consequent downregulation of stress-response signaling pathways may be key adaptation mechanisms that mediate the effects of FASN on cancer cell survival and metastasis, providing a strong rationale for targeting this pathway in advanced CRC.


Subject(s)
Colorectal Neoplasms/metabolism , Fatty Acid Synthase, Type I/biosynthesis , Cell Line, Tumor , Cell Respiration/physiology , Cell Survival/physiology , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Fatty Acid Synthase, Type I/antagonists & inhibitors , Fatty Acid Synthase, Type I/metabolism , Glycolysis , HCT116 Cells , HT29 Cells , Humans , Signal Transduction , Transcriptional Activation , Up-Regulation
14.
J Mol Cell Cardiol ; 72: 1-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24560668

ABSTRACT

Heart failure is associated with pump dysfunction and remodeling but it is not yet known if the condition affects different transmural regions of the heart in the same way. We tested the hypotheses that the left ventricles of non-failing human hearts exhibit transmural heterogeneity of cellular level contractile properties, and that heart failure produces transmural region-specific changes in contractile function. Permeabilized samples were prepared from the sub-epicardial, mid-myocardial, and sub-endocardial regions of the left ventricular free wall of non-failing (n=6) and failing (n=10) human hearts. Power, an in vitro index of systolic function, was higher in non-failing mid-myocardial samples (0.59±0.06µWmg(-1)) than in samples from the sub-epicardium (p=0.021) and the sub-endocardium (p=0.015). Non-failing mid-myocardial samples also produced more isometric force (14.3±1.33kNm(-2)) than samples from the sub-epicardium (p=0.008) and the sub-endocardium (p=0.026). Heart failure reduced power (p=0.009) and force (p=0.042) but affected the mid-myocardium more than the other transmural regions. Fibrosis increased with heart failure (p=0.021) and mid-myocardial tissue from failing hearts contained more collagen than matched sub-epicardial (p<0.001) and sub-endocardial (p=0.043) samples. Power output was correlated with the relative content of actin and troponin I, and was also statistically linked to the relative content and phosphorylation of desmin and myosin light chain-1. Non-failing human hearts exhibit transmural heterogeneity of contractile properties. In failing organs, region-specific fibrosis produces the greatest contractile deficits in the mid-myocardium. Targeting fibrosis and sarcomeric proteins in the mid-myocardium may be particularly effective therapies for heart failure.


Subject(s)
Endocardium/physiopathology , Heart Failure/physiopathology , Heart Ventricles/physiopathology , Myocardium/pathology , Pericardium/physiopathology , Actins/genetics , Actins/metabolism , Adolescent , Adult , Aged , Desmin/genetics , Desmin/metabolism , Endocardium/metabolism , Female , Fibrosis , Gene Expression , Heart Failure/metabolism , Heart Failure/surgery , Heart Transplantation , Heart Ventricles/metabolism , Humans , Isometric Contraction , Male , Middle Aged , Myocardial Contraction , Myocardium/metabolism , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Organ Specificity , Pericardium/metabolism , Troponin I/genetics , Troponin I/metabolism
15.
Arch Biochem Biophys ; 552-553: 92-9, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24012810

ABSTRACT

Diastolic dysfunction is a clinically significant problem for patients with diabetes and often reflects increased ventricular stiffness. Attached cross-bridges contribute to myocardial stiffness and produce short-range forces, but it is not yet known whether these forces are altered in diabetes. In this study, we tested the hypothesis that cross-bridge-based short-range forces are increased in the streptozotocin (STZ) induced rat model of type 1 diabetes. Chemically permeabilized myocardial preparations were obtained from 12week old rats that had been injected with STZ or vehicle 4weeks earlier, and activated in solutions with pCa (=-log10[Ca(2+)]) values ranging from 9.0 to 4.5. The short-range forces elicited by controlled length changes were ∼67% greater in the samples from the diabetic rats than in the control preparations. This change was mostly due to an increased elastic limit (the length change at the peak short-range force) as opposed to increased passive muscle stiffness. The STZ-induced increase in short-ranges forces is thus unlikely to reflect changes to titin and/or collagen filaments. Gel electrophoresis showed that STZ increased the relative expression of ß myosin heavy chain. This molecular mechanism can explain the increased short-ranges forces observed in the diabetic tissue if ß myosin molecules remain bound between the filaments for longer durations than α molecules during imposed movements. These results suggest that interventions that decrease myosin attachment times may be useful treatments for diastolic dysfunction associated with diabetes.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Myocardium/metabolism , Myocardium/pathology , Myosin Heavy Chains/metabolism , Ventricular Myosins/metabolism , Animals , Biomechanical Phenomena , Female , Male , Myosin Heavy Chains/analysis , Rats , Rats, Sprague-Dawley , Ventricular Myosins/analysis
16.
Neurotox Res ; 22(3): 220-30, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22083458

ABSTRACT

Recent studies have demonstrated the re-emergence of cell cycle proteins in brain as patients progress from the early stages of mild cognitive impairment (MCI) into Alzheimer's disease (AD). Oxidative stress markers present in AD have also been shown to be present in MCI brain suggesting that these events occur in early stages of the disease. The levels of key cell cycle proteins, such as CDK2, CDK5, cyclin G1, and BRAC1 have all been found to be elevated in MCI brain compared to age-matched control. Further, peptidyl prolyl cis-trans isomerase (Pin1), a protein that plays an important role in regulating the activity of key proteins, such as CDK5, GSK3-ß, and PP2A that are involved in both the phosphorylation state of Tau and in the cell cycle, has been found to be oxidatively modified and downregulated in both AD and MCI brain. Hyperphosphorylation of Tau then results in synapse loss and the characteristic Tau aggregation as neurofibrillary tangles, an AD hallmark. In this review, we summarized the role of cell cycle dysregulation in the progression of disease from MCI to AD. Based on the current literature, it is tempting to speculate that a combination of oxidative stress and cell cycle dysfunction conceivably leads to neurodegeneration.


Subject(s)
Alzheimer Disease/etiology , Brain/metabolism , Cell Cycle Proteins/metabolism , Cognitive Dysfunction/pathology , Cognitive Dysfunction/physiopathology , Disease Progression , Animals , Humans
17.
Electrophoresis ; 30(5): 848-51, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19197901

ABSTRACT

GelBandFitter is a computer program that uses non-linear regression techniques to fit mathematical functions to densitometry profiles of protein gels. This allows for improved quantification of gels with partially overlapping and potentially asymmetric protein bands. The program can also be used to analyze immunoblots with closely spaced bands. GelBandFitter was developed in Matlab and the source code and/or a Windows executable file can be downloaded at no cost to academic users from http://www.gelbandfitter.org.


Subject(s)
Electrophoresis , Immunoblotting , Nonlinear Dynamics , Software , Animals , Cardiac Myosins/isolation & purification , Myosin Heavy Chains/isolation & purification , Rats , User-Computer Interface
18.
J Mol Cell Cardiol ; 46(1): 39-46, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19007786

ABSTRACT

The mechanical properties of triton-permeabilized ventricular preparations isolated from 4, 18 and 24-month-old F344 rats were analyzed to provide information about the molecular mechanisms that lead to age-related increases in diastolic myocardial stiffness in these animals. Passive stiffness (measured in solutions with minimal free Ca(2+)) did not change with age. This implies that the aging-associated dysfunction is not due to changes in titin or collagen molecules. Ca(2+)-activated preparations exhibited a characteristic short-range force response: force rose rapidly until the muscle reached its elastic limit and less rapidly thereafter. The elastic limit increased from 0.43+/-0.01% l(0) (where l(0) is the initial muscle length) in preparations from 4-month-old animals to 0.49+/-0.01% l(0) in preparations from 24-month-old rats (p<0.001, ANOVA). Relative short-range force was defined as the maximum force produced during the short-range response normalized to the prevailing tension. This parameter increased from 0.110+/-0.002 to 0.142+/-0.002 over the same age-span (p<0.001, ANOVA). Analytical gel electrophoresis showed that the maximum stiffness of the preparations during the short-range response and the relative short-range force increased (p=0.031 and p=0.005 respectively) with the relative content of slow beta myosin heavy chain molecules. Elastic limit values did not correlate with myosin isoform content. Simulations based on these results suggest that attached beta myosin heavy chain cross-bridges are stiffer than links formed by alpha myosin heads. In conclusion, elevated content of stiffer beta myosin heavy chain molecules may contribute to aging-associated increases in myocardial stiffness.


Subject(s)
Myocardial Contraction/physiology , Myocardium/pathology , Aging , Algorithms , Animals , Calcium/metabolism , Detergents/pharmacology , Female , Models, Theoretical , Muscles/pathology , Myosin Heavy Chains/metabolism , Protein Isoforms , Rats , Rats, Inbred F344 , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...