Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Int J Mol Sci ; 24(8)2023 Apr 16.
Article in English | MEDLINE | ID: mdl-37108509

ABSTRACT

By 2050, at least 700 million people will require hearing therapy while 2.5 billion are projected to suffer from hearing loss. Sensorineural hearing loss (SNHL) arises from the inability of the inner ear to convert fluid waves into neural electric signals because of injury to cochlear hair cells that has resulted in their death. In addition, systemic chronic inflammation implicated in other pathologies may exacerbate cell death leading to SNHL. Phytochemicals have emerged as a possible solution because of the growing evidence of their anti-inflammatory, antioxidant, and anti-apoptotic properties. Ginseng and its bioactive molecules, ginsenosides, exhibit effects that suppress pro-inflammatory signaling and protect against apoptosis. In the current study, we investigated the effects of ginsenoside Rc (G-Rc) on UB/OC-2 primary murine sensory hair cell survival in response to palmitate-induced injury. G-Rc promoted UB/OC-2 cell survival and cell cycle progression. Additionally, G-Rc enhanced the differentiation of UB/OC-2 cells into functional sensory hair cells and alleviated palmitate-induced inflammation, endoplasmic reticulum stress, and apoptosis. The current study offers novel insights into the effects of G-Rc as a potential adjuvant for SNHL and warrants further studies elucidating the molecular mechanisms.


Subject(s)
Ginsenosides , Hearing Loss, Sensorineural , Panax , Humans , Mice , Animals , Ginsenosides/pharmacology , Panax/chemistry , Cochlea , Inflammation
2.
Cell Commun Signal ; 18(1): 126, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32795297

ABSTRACT

BACKGROUND: Current pharmacological therapies and treatments targeting pancreatic neuroendocrine tumors (PNETs) have proven ineffective, far too often. Therefore, there is an urgent need for alternative therapeutic approaches. Zyflamend, a combination of anti-inflammatory herbal extracts, that has proven to be effective in various in vitro and in vivo cancer platforms, shows promise. However, its effects on pancreatic cancer, in particular, remain largely unexplored. METHODS: In the current study, we investigated the effects of Zyflamend on the survival of beta-TC-6 pancreatic insulinoma cells (ß-TC6) and conducted a detailed analysis of the underlying molecular mechanisms. RESULTS: Herein, we demonstrate that Zyflamend treatment decreased cell proliferation in a dose-dependent manner, concomitant with increased apoptotic cell death and cell cycle arrest at the G2/M phase. At the molecular level, treatment with Zyflamend led to the induction of ER stress, autophagy, and the activation of c-Jun N-terminal kinase (JNK) pathway. Notably, pharmacological inhibition of JNK abrogated the pro-apoptotic effects of Zyflamend. Furthermore, Zyflamend exacerbated the effects of streptozotocin and adriamycin-induced ER stress, autophagy, and apoptosis. CONCLUSION: The current study identifies Zyflamend as a potential novel adjuvant in the treatment of pancreatic cancer via modulation of the JNK pathway. Video abstract.


Subject(s)
Apoptosis , MAP Kinase Signaling System , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Plant Extracts/pharmacology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/pharmacology , Endoplasmic Reticulum Stress/drug effects , Inflammation/pathology , MAP Kinase Signaling System/drug effects , Mice , Models, Biological , Rats , Streptozocin/pharmacology
3.
Adipocyte ; 9(1): 454-471, 2020 12.
Article in English | MEDLINE | ID: mdl-32779962

ABSTRACT

The prevalence of obesity and its comorbidities has sparked a worldwide concern to address rates of adipose tissue accrual. Recent studies have demonstrated a novel role of Zyflamend, a blend of natural herbal extracts, in regulating lipid metabolism in several cancer cell lines through the activation of the AMPK signalling pathway. Yet, the role of Zyflamend in adipogenic differentiation and lipid metabolism remains largely unexplored. The objective of this study is to investigate the effects of Zyflamend on white 3T3-MBX pre-adipocyte differentiation and elucidate the molecular mechanisms. We demonstrate that Zyflamend treatment altered cell cycle progression, attenuated proliferation, and increased cell death of 3T3-MBX pre-adipocytes. In addition, treatment with Zyflamend inhibited lipid accumulation during the differentiation of 3T3-MBX cells, consistent with decreased expression of lipogenic genes and increased lipolysis. Mechanistically, Zyflamend-induced alterations in adipogenesis were mediated, at least in part, through the activation of AMPK, PKA, and JNK. Inhibition of AMPK partially reversed Zyflamend-induced inhibition of differentiation, whereas the inhibition of either JNK or PKA fully restored adipocyte differentiation and decreased lipolysis. Taken together, the present study demonstrates that Zyflamend, as a novel anti-adipogenic bioactive mix, inhibits adipocyte differentiation through the activation of the PKA and JNK pathways. ABBREVIATION: 7-AAD: 7-amino-actinomycin D; ACC: acetyl-CoA carboxylase; AKT: protein kinase B; AMPK: AMP-activated protein kinase; ATGL: adipose triglyceride lipase; C/EBPα: CCAAT-enhancer binding protein alpha; DMEM: Dulbecco's Modified Eagle Medium; DMSO: dimethyl sulphoxide; DTT: dithiothreitol; EGTA: ethylene glycol-bis-(2-aminoethyl)-N,N,N',N'-tetraacetic acid; ERK: extracellular signal-regulated kinases; FASN: fatty acid synthase; FBS: foetal bovine serum; GLUT: glucose transporter; HSL: hormone-sensitive lipase; IR: insulin receptor; IRS: insulin receptor substrate; JNK: c-JUN N-terminal kinase; MGL: monoacylglycerol lipase; NaF: sodium fluoride; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; PBS: phosphate buffered- saline; PCB: pyruvate carboxylase; PDE: phosphodiesterase; PKA: protein kinase cAMP-dependent; PMSF: phenylmethylsulfonyl fluoride; PPARγ: perilipin peroxisome proliferator-activated receptor gamma; PREF-1: pre-adipocyte factor 1; PVDF: polyvinylidene fluoride; RIPA: radio-immunoprecipitation assay; SDS-PAGE: sodium dodecyl sulphate polyacrylamide gel electrophoresis; SEM: standard error of the mean; SOX9: suppressor of cytokine signalling 9; TGs: triacylglycerols.


Subject(s)
Adipogenesis/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Plant Extracts/pharmacology , Signal Transduction/drug effects , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Lipolysis , Mice
4.
Article in English | MEDLINE | ID: mdl-32485595

ABSTRACT

The purpose of this research was to explore various allometric scaling models for dietary nutrients to improve translational validity between preclinical experimental rodent models and humans, focusing on polyunsaturated fats. Currently, there is no authoritative document that provides standardized guidelines for which dietary designs can be based on to improve translational fidelity between species. This paper reviews the challenges of using a rodent model, the major allometric scaling models, the use of these mathematical models to extrapolate human equivalent doses, and then tests one of these models using data generated in mice, with comparisons of data generated in human clinical trials. Mice were fed diets containing micro- and macronutrient compositions that approximated the US diet based on energy distribution and were then supplemented with increasing levels of various n-3 and n-6 polyunsaturated fatty acids at human equivalent doses. Changes in plasma and erythrocyte fatty acid phospholipid compositions were determined and compared to corresponding data generated in humans. Our findings suggest that basing lipid composition on percent of energy may result in comparable outcomes between mice and humans and that extrapolation of non-energy producing nutrients between species might be done using differences in energy needs (based on food intake).


Subject(s)
Dietary Fats, Unsaturated/administration & dosage , Erythrocytes/chemistry , Fatty Acids, Unsaturated/blood , Plasma/chemistry , Animals , Dietary Fats, Unsaturated/pharmacokinetics , Guidelines as Topic , Humans , Mice , Models, Animal , Rats , Translational Research, Biomedical
5.
Free Radic Biol Med ; 143: 176-192, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31401304

ABSTRACT

Pyruvate kinase M2 is a critical enzyme that regulates cell metabolism and growth under different physiological conditions. In its metabolic role, pyruvate kinase M2 catalyzes the last glycolytic step which converts phosphoenolpyruvate to pyruvate with the generation of ATP. Beyond this metabolic role in glycolysis, PKM2 regulates gene expression in the nucleus, phosphorylates several essential proteins that regulate major cell signaling pathways, and contribute to the redox homeostasis of cancer cells. The expression of PKM2 has been demonstrated to be significantly elevated in several types of cancer, and the overall inflammatory response. The unusual pattern of PKM2 expression inspired scientists to investigate the unrevealed functions of PKM2 and the therapeutic potential of targeting PKM2 in cancer and other disorders. Therefore, the purpose of this review is to discuss the mechanistic and therapeutic potential of targeting PKM2 with the focus on cancer metabolism, redox homeostasis, inflammation, and metabolic disorders. This review highlights and provides insight into the metabolic and non-metabolic functions of PKM2 and its relevant association with health and disease.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic , Inflammation/enzymology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Metabolic Diseases/enzymology , Thyroid Hormones/metabolism , Adenosine Triphosphate/metabolism , Atherosclerosis/enzymology , Cell Proliferation , Glycolysis , Homeostasis , Humans , Inflammatory Bowel Diseases/enzymology , Insulin/metabolism , Kidney Diseases/enzymology , Liver/enzymology , Naphthoquinones/pharmacology , Neoplasm Metastasis , Neoplasms/enzymology , Neuralgia/enzymology , Oxidants/metabolism , Oxidation-Reduction , Protein Isoforms , Sepsis/enzymology , Signal Transduction , Tissue Distribution , Thyroid Hormone-Binding Proteins
6.
Nutr Res ; 68: 34-44, 2019 08.
Article in English | MEDLINE | ID: mdl-31306903

ABSTRACT

The polyherbal blend Zyflamend™ has been shown to have anti-inflammatory properties and attenuate inflammatory-modulated pathologies. Fish oils have also been shown to have cardioprotective properties. However, the beneficial effects of their combination have not been investigated. Intimal hyperplasia (IH), a pathological remodeling response of a vessel to injury, is heavily regulated by an immune-mediated reaction. The objective of this study was to determine if dietary supplementation with Zyflamend and/or Wholemega could affect inflammatory-dependent vascular remodeling mechanisms when provided at human equivalent doses. Based on their anti-inflammatory properties and protective benefits demonstrated in previous pre-clinical studies, we hypothesized administration of these supplements would prevent IH in an animal model of vascular injury. The diets of aged male rats were supplemented with human equivalent doses of Zyflamend (Zyf) and/or Wholemega (WMega) or placebo (Plac) for 1wk prior to balloon angioplasty (BA)-induced injury of the left carotid artery. At 28d post-injury morphometric analysis of carotid tissue revealed IH was decreased in Zyf + WMega animals compared to placebo, while Zyf or WMega independently had no significant effect. Serum cytokine screening indicated injury-induced interleukin family isoforms, interferon-γ, and macrophage inflammatory proteins were downregulated by Zyf + WMega. Immunohistochemical staining for monocyte/macrophage phenotypic markers revealed that while overall monocyte/macrophage vessel infiltration was not affected, Zyf + WMega limited the alternative differentiation of M2 macrophages and reduced the presence of myofibroblasts in the injured vessel wall. In summary, dietary supplementation with Zyf + WMega attenuated the acute inflammatory response following vascular injury and inhibited IH development in vivo.


Subject(s)
Carotid Artery Injuries/pathology , Carotid Artery, Common/pathology , Fish Oils/administration & dosage , Plant Extracts/administration & dosage , Angioplasty, Balloon , Animals , Carotid Artery Injuries/etiology , Carotid Artery, Common/chemistry , Cytokines/blood , Diet , Dietary Supplements , Female , Hyperplasia/prevention & control , Inflammation/blood , Male , Placebos , Rats , Rats, Sprague-Dawley
7.
Oncotarget ; 9(43): 27280-27292, 2018 Jun 05.
Article in English | MEDLINE | ID: mdl-29930765

ABSTRACT

Colorectal cancer is characterized by an increase in the utilization of glucose and a diminishment in the oxidation of butyrate, which is a short chain fatty acid. In colorectal cancer cells, butyrate inhibits histone deacetylases to increase the expression of genes that slow the cell cycle and induce apoptosis. Understanding the mechanisms that contribute to the metabolic shift away from butyrate oxidation in cancer cells is important in in understanding the beneficial effects of the molecule toward colorectal cancer. Here, we demonstrate that butyrate decreased its own oxidation in cancerous colonocytes. Butyrate lowered the expression of short chain acyl-CoA dehydrogenase, an enzyme that mediates the oxidation of short-chain fatty acids. Butyrate does not alter short chain acyl-CoA dehydrogenase levels in non-cancerous colonocytes. Trichostatin A, a structurally unrelated inhibitor of histone deacetylases, and propionate also decreased the level of short chain acyl-CoA dehydrogenase, which alluded to inhibition of histone deacetylases as a part of the mechanism. Knockdown of histone deacetylase isoform 1, but not isoform 2 or 3, inhibited the ability of butyrate to decrease short chain acyl-CoA dehydrogenase expression. This work identifies a mechanism by which butyrate selective targets colorectal cancer cells to reduce its own metabolism.

8.
BMC Complement Altern Med ; 18(1): 188, 2018 Jun 18.
Article in English | MEDLINE | ID: mdl-29914450

ABSTRACT

BACKGROUND: Zyflamend, a blend of herbal extracts, effectively inhibits tumor growth using preclinical models of castrate-resistant prostate cancer mediated in part by 5'-adenosine monophosphate-activated protein kinase (AMPK), a master energy sensor of the cell. Clinically, treatment with Zyflamend and/or metformin (activators of AMPK) had benefits in castrate-resistant prostate cancer patients who no longer responded to treatment. Two predominant upstream kinases are known to activate AMPK: liver kinase B1 (LKB1), a tumor suppressor, and calcium-calmodulin kinase kinase-2 (CaMKK2), a tumor promotor over-expressed in many cancers. The objective was to interrogate how Zyflamend activates AMPK by determining the roles of LKB1 and CaMKK2. METHODS: AMPK activation was determined in CWR22Rv1 cells treated with a variety of inhibitors of LKB1 and CaMKK2 in the presence and absence of Zyflamend, and in LKB1-null HeLa cells that constitutively express CaMKK2, following transfection with wild type LKB1 or catalytically-dead mutants. Upstream regulation by Zyflamend of LKB1 and CaMKK2 was investigated targeting protein kinase C-zeta (PKCζ) and death-associated protein kinase (DAPK), respectively. RESULTS: Zyflamend's activation of AMPK appears to be LKB1 dependent, while simultaneously inhibiting CaMKK2 activity. Zyflamend failed to rescue the activation of AMPK in the presence of pharmacological and molecular inhibitors of LKB1, an effect not observed in the presence of inhibitors of CaMKK2. Using LKB1-null and catalytically-dead LKB1-transfected HeLa cells that constitutively express CaMKK2, ionomycin (activator of CaMKK2) increased phosphorylation of AMPK, but Zyflamend only had an effect in cells transfected with wild type LKB1. Zyflamend appears to inhibit CaMKK2 by DAPK-mediated phosphorylation of CaMKK2 at Ser511, an effect prevented by a DAPK inhibitor. Alternatively, Zyflamend mediates LKB1 activation via increased phosphorylation of PKCζ, where it induced translocation of PKCζ and LKB1 to their respective active compartments in HeLa cells following treatment. Altering the catalytic activity of LKB1 did not alter this translocation. DISCUSSION: Zyflamend's activation of AMPK is mediated by LKB1, possibly via PKCζ, but independent of CaMKK2 by a mechanism that appears to involve DAPK. CONCLUSIONS: Therefore, this is the first evidence that natural products simultaneously and antithetically regulate upstream kinases, known to be involved in cancer, via the activation of AMPK.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Plant Extracts/pharmacology , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Cell Line, Tumor , HeLa Cells , Humans , Male , Phosphorylation/drug effects , Signal Transduction/drug effects
9.
J Med Food ; 21(3): 306-316, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29227176

ABSTRACT

Zyflamend is a highly controlled blend of 10 herbal extracts that synergistically impact multiple cell signaling pathways with anticancer and anti-inflammatory properties. More recently, its effects were shown to also modify cellular energetics, for example, activation of fatty acid oxidation and inhibition of lipogenesis. However, its general metabolic effects in vivo have yet to be explored. The objective of this study was to characterize the tissue specific metabolomes in response to supplementation of Zyflamend in mice, with a comparison of equivalent metabolomics data generated in plasma from humans supplemented with Zyflamend. Because Zyflamend has been shown to activate AMPK, the "energy sensor" of the cell, in vitro, the effects of Zyflamend on adiposity were also tested in the murine model. C57BL/6 mice were fed diets that mimicked the macro- and micronutrient composition of the U.S. diet with and without Zyflamend supplementation at human equivalent doses. Untargeted metabolomics was performed in liver, skeletal muscle, adipose, and plasma from mice consuming Zyflamend and in plasma from humans supplemented with Zyflamend at an equivalent dose. Adiposity in mice was significantly reduced in the Zyflamend-treated animals (compared with controls) without affecting body weight or weight gain. Based on KEGG pathway enrichment, purine and pyrimidine metabolism (potential regulators of AMPK) were particularly responsive to Zyflamend across all tissues, but only in mice. Consistent with the metabolomics data, Zyflamend activated AMPK and inhibited acetyl CoA-carboxylase in adipose tissue, key regulators of lipogenesis. Zyflamend reduces adipose tissue in mice through a mechanism that likely involves the activation of AMPK.


Subject(s)
Abdominal Fat/metabolism , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Dietary Supplements , Liver/metabolism , Muscle, Skeletal/metabolism , Plant Extracts/administration & dosage , Abdominal Fat/enzymology , Adiposity , Adult , Aged , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Antineoplastic Agents, Phytogenic/adverse effects , Biomarkers/blood , Biomarkers/metabolism , Dietary Supplements/adverse effects , Discriminant Analysis , Energy Metabolism , Humans , Liver/enzymology , Male , Metabolomics/methods , Mice, Inbred C57BL , Middle Aged , Muscle, Skeletal/enzymology , Organ Specificity , Plant Extracts/adverse effects , Principal Component Analysis , Random Allocation , Species Specificity
10.
J Cell Biochem ; 118(6): 1614-1621, 2017 06.
Article in English | MEDLINE | ID: mdl-27922186

ABSTRACT

Cancer, in part, is driven, by alterations in cellular metabolism that promote cell survival and cell proliferation. Identifying factors that influence this shift in cellular metabolism in cancer cells is important. Interleukin-1ß (IL-1ß) is a pro-inflammatory cytokine that has been reported to be elevated in colorectal cancer patients. While much is known toward the effect of dietary nutrients on regulating inflammation and the inflammatory response, which includes cytokines such as IL-1ß, far less is understood how cytokines impact nutrient fate to alter cancer cell metabolism. Butyrate, a nutrient derived from the fermentation of dietary fiber in the colon, is the preferential exogenous energetic substrate used by non-cancerous colonocytes, but is used less efficiently by colorectal cancer cells. To test whether IL-1ß alters colonocyte energy metabolism, we measured butyrate oxidation in HCT116 colorectal cancer cells with and without IL-1ß. We hypothesize that IL-1ß will push cancerous colonocytes away from the utilization and oxidation of butyrate. In this study, we demonstrate that pretreatment of colorectal cancer cells with IL-1ß diminished butyrate oxidation and NADH levels. This effect was blocked with the interleukin receptor antagonist A (IL-1RA). Moreover, IL-1ß suppressed basal mitochondrial respiration and lowered the mitochondrial spare capacity. By using inhibitors to block downstream targets of the interleukin-1 receptor pathway, we show that p38 is required for the IL-1ß-mediated decrease in butyrate oxidation. These data provide insight into the metabolic effects induced by IL-1ß in colorectal cancer, and identify relevant targets that may be exploited to block the effects of this cytokine. J. Cell. Biochem. 118: 1614-1621, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Butyric Acid/metabolism , Colorectal Neoplasms/metabolism , Glucose/metabolism , Interleukin-1beta/metabolism , Energy Metabolism , HCT116 Cells , Humans , Mitochondria/metabolism , NAD/metabolism , Oxidation-Reduction
11.
J Am Coll Nutr ; 35(5): 452-68, 2016 07.
Article in English | MEDLINE | ID: mdl-26595854

ABSTRACT

Animal studies have shown that dietary omega-3 polyunsaturated fatty acids (n-3) may play a role in the development of prostate cancer, but the results of epidemiologic studies have been equivocal. Associations in humans may vary depending on study design, measurement methodology of fatty acid intake, intake ranges, and stage of cancer development. To address this, we identified 36 published studies through PubMed (Medline) from 1993 through 2013 on long-chain n-3s and prostate cancer. Exposure measurements included dietary assessment and biomarker levels. Associations for total, early, and late stage prostate cancer were examined by subgroup of study design and exposure measure type and by using forest plots to illustrate the relative strength of associations within each subgroup. We also tested for potential threshold effects by considering studies that included measurement cut-points that met intake levels recommended by the American Heart Association. We found no consistent evidence supporting a role of n-3s in either the causation or prevention of prostate cancer at any stage or grade. Results did not vary appreciably by study design, exposure measurement, intake level, or stage of cancer development.


Subject(s)
Diet , Fatty Acids, Omega-3/administration & dosage , Prostatic Neoplasms , Case-Control Studies , Cohort Studies , Docosahexaenoic Acids/administration & dosage , Eicosapentaenoic Acid/administration & dosage , Humans , MEDLINE , Male , Prostatic Neoplasms/chemically induced , Prostatic Neoplasms/pathology , Prostatic Neoplasms/prevention & control , Risk Factors , alpha-Linolenic Acid/administration & dosage
12.
J Cell Physiol ; 231(8): 1804-13, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26661480

ABSTRACT

Dietary fiber has been suggested to suppress colorectal cancer development, although the mechanisms contributing to this beneficial effect remain elusive. Butyrate, a fermentation product of fiber, has been shown to have anti-proliferative and pro-apoptotic effects on colorectal cancer cells. The metabolic fate of butyrate in the cell is important in determining whether, it acts as an HDAC inhibitor or is consumed as a short-chain fatty acid. Non-cancerous colonocytes utilize butyrate as the primary energy source whereas cancerous colonocytes increase glucose utilization through the Warburg effect. In this study, we show that butyrate oxidation is decreased in cancerous colonocytes compared to non-cancerous colonocytes. We demonstrate that colorectal cancer cells utilize both a carnitine-dependent and carnitine-independent mechanism that contributes to butyrate oxidation. The carnitine-dependent mechanism is contingent on butyrate concentration. Knockdown of CPT1A in colorectal cancer cells abolishes butyrate oxidation. In terms of selectivity, the carnitine-dependent mechanism only regulated butyrate oxidation, as acetate and propionate oxidation were carnitine-independent. Carnitine decreased the action of butyrate as an HDAC inhibitor and suppressed induction of H3 acetylation by butyrate in colorectal cancer cells. Thus, diminished oxidation of butyrate is associated with decreased HDAC inhibition and histone acetylation. In relation to the mechanism, we find that dichloroacetate, which decreases phosphorylation of pyruvate dehydrogenase, increased butyrate oxidation and that this effect was carnitine-dependent. In conclusion, these data suggest that colorectal cancer cells decrease butyrate oxidation through inhibition of pyruvate dehydrogenase, which is carnitine-dependent, and provide insight into why butyrate shows selective effects toward colorectal cancer cells. J. Cell. Physiol. 231: 1804-1813, 2016. © 2015 Wiley Periodicals, Inc.


Subject(s)
Antineoplastic Agents/pharmacology , Butyric Acid/pharmacology , Carnitine/metabolism , Colorectal Neoplasms/drug therapy , Energy Metabolism/drug effects , Histone Deacetylase Inhibitors/pharmacology , Acetylation , Antineoplastic Agents/metabolism , Butyric Acid/metabolism , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Dichloroacetic Acid/pharmacology , Dose-Response Relationship, Drug , HCT116 Cells , Histone Deacetylase Inhibitors/metabolism , Histones/metabolism , Humans , Organic Cation Transport Proteins/metabolism , Oxidation-Reduction , Phosphorylation , Pyruvate Dehydrogenase Complex/antagonists & inhibitors , Pyruvate Dehydrogenase Complex/metabolism , RNA Interference , Signal Transduction/drug effects , Solute Carrier Family 22 Member 5 , Time Factors , Transfection
13.
Cancer Prev Res (Phila) ; 8(10): 989-99, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26290393

ABSTRACT

Current therapies for treatment of myeloid leukemia do not eliminate leukemia stem cells (LSC), leading to disease relapse. In this study, we supplemented mice with eicosapentaenoic acid (EPA, C20:5), a polyunsaturated omega-3 fatty acid, at pharmacologic levels, to examine whether the endogenous metabolite, cyclopentenone prostaglandin delta-12 PGJ3 (Δ(12)-PGJ3), was effective in targeting LSCs in experimental leukemia. EPA supplementation for 8 weeks resulted in enhanced endogenous production of Δ(12)-PGJ3 that was blocked by indomethacin, a cyclooxygenase (COX) inhibitor. Using a murine model of chronic myelogenous leukemia (CML) induced by bone marrow transplantation of BCR-ABL-expressing hematopoietic stem cells, mice supplemented with EPA showed a decrease in the LSC population, and reduced splenomegaly and leukocytosis, when compared with mice on an oleic acid diet. Supplementation of CML mice carrying the T315I mutation (in BCR-ABL) with EPA resulted in a similar effect. Indomethacin blocked the EPA effect and increased the severity of BCR-ABL-induced CML and decreased apoptosis. Δ(12)-PGJ3 rescued indomethacin-treated BCR-ABL mice and decreased LSCs. Inhibition of hematopoietic-prostaglandin D synthase (H-PGDS) by HQL-79 in EPA-supplemented CML mice also blocked the effect of EPA. In addition, EPA supplementation was effective in a murine model of acute myeloid leukemia. EPA-supplemented mice exhibited a decrease in leukemia burden and a decrease in the LSC colony-forming unit (LSC-CFU). The decrease in LSCs was confirmed through serial transplantation assays in all disease models. The results support a chemopreventive role for EPA in myeloid leukemia, which is dependent on the ability to efficiently convert EPA to endogenous COX-derived prostanoids, including Δ(12)-PGJ3.


Subject(s)
Anticarcinogenic Agents/pharmacology , Dietary Supplements , Eicosapentaenoic Acid/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/drug effects , Animals , Chromatography, Liquid , Flow Cytometry , HEK293 Cells , Humans , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Tandem Mass Spectrometry , Transduction, Genetic , Xenograft Model Antitumor Assays
14.
Nutr Res ; 35(4): 328-36, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25640963

ABSTRACT

Type 1 diabetes mellitus results from autoimmune-mediated destruction of pancreatic islet ß-cells, a process associated with inflammatory signals. We hypothesized that dietary supplementation with botanicals known to contain anti-inflammatory properties would prevent losses in functional ß-cell mass in nonobese diabetic (NOD) mice, a rodent model of autoimmune-mediated islet inflammation that spontaneously develops diabetes. Female NOD mice, a model of spontaneous autoimmune diabetes, were fed a diet supplemented with herbal extracts (1.916 g total botanical extracts per 1 kg of diet) over a 12-week period. The mice consumed isocaloric matched diets without (controls) and with polyherbal supplementation (PHS) ad libitum starting at a prediabetic stage (age 6 weeks) for 12 weeks. Control mice developed hyperglycemia (>180 mg/dL) within 16 weeks (n = 9). By contrast, mice receiving the PHS diet did not develop hyperglycemia by 18 weeks (n = 8). Insulin-positive cell mass within pancreatic islets was 31.9% greater in PHS mice relative to controls. We also detected a 26% decrease in CD3(+) lymphocytic infiltration in PHS mice relative to mice consuming a control diet. In vitro assays revealed reduced ß-cell expression of the chemokines CCL2 and CXCL10 after overnight PHS addition to the culture media. We conclude that dietary PHS delays initiation of autoimmune-mediated ß-cell destruction and subsequent onset of diabetes mellitus by diminishing islet inflammatory responses.


Subject(s)
Dietary Supplements , Hyperglycemia/drug therapy , Insulin-Secreting Cells/drug effects , Plant Extracts/pharmacology , Animals , Cell Line , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Diet/veterinary , Female , Inflammation Mediators/pharmacology , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred NOD , Prediabetic State/drug therapy , Rats
15.
BMC Complement Altern Med ; 14: 68, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24555771

ABSTRACT

BACKGROUND: Zyflamend, a mixture containing extracts of ten herbs, has shown promise in a variety of preclinical cancer models, including prostate cancer. The current experiments were designed to investigate the effects of Zyflamend on the expression of class I and II histone deacetylases, a family of enzymes known to be over expressed in a variety of cancers. METHODS: CWR22Rv1 cells, a castrate-resistant prostate cancer cell line, were treated with Zyflamend and the expression of class I and II histone deacetylases, along with their downstream target the tumor suppressor gene p21, was investigated. Involvement of p21 was confirmed with siRNA knockdown and over expression experiments. RESULTS: Zyflamend down-regulated the expression of all class I and II histone deacetylases where Chinese goldthread and baikal skullcap (two of its components) appear to be primarily responsible for these results. In addition, Zyflamend up regulated the histone acetyl transferase complex CBP/p300, potentially contributing to the increase in histone 3 acetylation. Expression of the tumor suppressor gene p21, a known downstream target of histone deacetylases and CBP/p300, was increased by Zyflamend treatment and the effect on p21 was, in part, mediated through Erk1/2. Knockdown of p21 with siRNA technology attenuated Zyflamend-induced growth inhibition. Over expression of p21 inhibited cell growth and concomitant treatment with Zyflamend enhanced this effect. CONCLUSIONS: Our results suggest that the extracts of this polyherbal combination increase histone 3 acetylation, inhibit the expression of class I and class II histone deacetylases, increase the activation of CBP/p300 and inhibit cell proliferation, in part, by up regulating p21 expression.


Subject(s)
Coptis , Histone Deacetylases/metabolism , Phytotherapy , Plant Extracts/pharmacology , Prostatic Neoplasms/metabolism , Scutellaria , Tumor Suppressor Proteins/metabolism , Acetylation , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Histones/metabolism , Humans , Male , Plant Extracts/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , RNA, Small Interfering/metabolism , Transcriptional Activation , Tumor Suppressor Proteins/drug effects , Up-Regulation , p300-CBP Transcription Factors/metabolism
16.
Biochem Cell Biol ; 91(6): 428-34, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24219284

ABSTRACT

Obesity is associated with chronic inflammation. Toll-like receptors (TLR) and NOD-like receptors (NLR) are two families of pattern recognition receptors that play important roles in the immune response and inflammation in adipocytes. Activation of TLR4 has been shown to stimulate lipolysis from adipose tissue or adipocytes. However, effects of activation of nucleotide-oligomerization domain containing protein 1 (NOD1), one of the prominent members of NLRs, on adipocyte lipolysis have not been studied. Here we report that NOD1 activation by the synthetic ligands (Tri-DAP and C12-iEDAP) stimulated lipolysis in 3T3-L1 adipocytes in a time- and dose-dependent manner. C12-iEDAP-induced lipolysis was attenuated with NOD1 siRNA knockdown, demonstrating the specificity of the effects. Moreover, inhibition of the protein kinase A (PKA)/hormone sensitive lipase (HSL) and NF-κB pathways by the pharmacological inhibitors attenuated the lipolytic effects of C12-iEDAP. Furthermore, we show NOD1 activation induced PKA activation independent of cAMP production and inhibition of NF-κB pathways attenuated phosphorylation of selected PKA lipolytic targets (phosphorylation of Perilipin Ser 517 and HSL Ser 563). Taken together, our results demonstrate a novel role of NOD1 activation, via NF-κB/PKA lipolytic activation, in inducing lipolysis in adipocytes and suggest that NOD1 activation may contribute to dyslipidemia in obesity.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Lipolysis/drug effects , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Signal Transduction/drug effects , 3T3-L1 Cells , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Diaminopimelic Acid/analogs & derivatives , Diaminopimelic Acid/pharmacology , Dose-Response Relationship, Drug , Gene Expression Regulation , Humans , Ligands , Mice , NF-kappa B/genetics , Nod1 Signaling Adaptor Protein/genetics , Oligopeptides/pharmacology , Perilipin-1 , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/genetics , Sterol Esterase/genetics , Sterol Esterase/metabolism
18.
Nutr Cancer ; 64(5): 749-60, 2012.
Article in English | MEDLINE | ID: mdl-22663543

ABSTRACT

Prostate cancer (PrC) is the second deadliest cancer of males in the United States Hormone deprivation therapy (HDT), a common therapy for advanced forms of the disease, results in tumor regression; unfortunately, tumors inevitably become castrate-resistant. Diet is not an appropriate primary therapy for refractory forms of the disease; however, diet may be effective as an adjuvant to HDT, potentially extending the latency period and delaying relapse and/or inhibiting refractory growth. Zyflamend® is a combination of extracts from multiple herbs, each with reported anticancer properties. Zyflamend can inhibit growth of various PrC cell lines, but no studies have investigated its potential use in vivo using a model of castrate-resistant PrC. In this study, oral doses of Zyflamend at human equivalent doses inhibited androgen-dependent and castrate-resistant tumor growth in a mouse model that mimics advanced stages of the disease, and reduced the expression of a number of biomarkers linked to PrC progression including pAKT, prostate specific antigen, histone deacetylases, and androgen receptor. In summary, this is the first article to report that Zyflamend, when provided at human equivalent doses, can potentiate the effects of hormone deprivation on tumor regression and growth inhibition of androgen-dependent and castrate-resistant PrC tumors in vivo.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Dietary Supplements , Plant Extracts/therapeutic use , Prostatic Neoplasms/diet therapy , Animals , Castration , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Male , Mice , Mice, Nude , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Testosterone/metabolism , Xenograft Model Antitumor Assays
19.
J Clin Lipidol ; 6(3): 216-34, 2012.
Article in English | MEDLINE | ID: mdl-22658146

ABSTRACT

Research dating back to the 1950s reported an association between the consumption of saturated fatty acids (SFAs) and risk of coronary heart disease. Recent epidemiological evidence, however, challenges these findings. It is well accepted that the consumption of SFAs increases low-density lipoprotein cholesterol (LDL-C), whereas carbohydrates, monounsaturated fatty acids (MUFAs), and polyunsaturated fatty acids (PUFAs) do not. High-density lipoprotein (HDL)-C increases with SFA intake. Among individuals who are insulin resistant, a low-fat, high-carbohydrate diet typically has an adverse effect on lipid profiles (in addition to decreasing HDL-C, it also increases triglyceride and LDL particle concentrations). Consequently, a moderate fat diet in which unsaturated fatty acids replace SFAs and carbohydrates are not augmented is advised to lower LDL-C; compared with a low-fat diet, a moderate-fat diet will lower triglycerides and increase HDL-C. Now, there is some new evidence that is questioning the health benefits of even MUFAs and PUFAs. In addition, in a few recent studies investigators have also failed to demonstrate expected cardiovascular benefits of marine-derived omega-3 fatty acids. To clarify the clinical pros and cons of dietary fats, the National Lipid Association held a fatty acid symposium at the 2011 National Lipid Association Scientific Sessions. During these sessions, the science regarding the effects of different fatty acid classes on coronary heart disease risk was reviewed.


Subject(s)
Coronary Disease/etiology , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Coronary Disease/diet therapy , Coronary Disease/prevention & control , Diet, Fat-Restricted , Dietary Carbohydrates , Dietary Fats/adverse effects , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Unsaturated , Humans , Risk Factors
20.
J Nutr ; 142(3): 641S-643S, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22323767

ABSTRACT

Faculty who had presented at the symposium "Heart Healthy Omega-3s (n-3 fatty acids) for Food: Stearidonic Acid (SDA) as a Sustainable Choice" met and agreed upon conclusions and recommendations that could be made on the basis of evidence provided at the symposium. The participants also submitted manuscripts relating to their topics and these are presented in this supplement. These manuscripts were reviewed and also contributed to the conclusions and recommendations presented herein. The three major objectives of the symposium were to: 1) increase understanding of the current and emerging knowledge regarding the health benefits of (n-3) fatty acids (FA) including a focus on stearidonic acid (SDA) and EPA; 2) evaluate the importance of increasing (n-3) FA consumption in the US and the current challenge of doing so via mainstream foods; and 3) consider the health and food application benefits of SDA as a precursor to EPA and a plant-based sustainable source of highly unsaturated (n-3) FA for mainstream foods. Specific areas for future research were defined and included in the summary and conclusions herein. Overall evidence-based conclusions included: the current evidence provides a strong rationale for increasing (n-3) FA intakes in the US and other populations; current consumption of (n-3) FA in most populations is either insufficient or not efficient at providing adequate tissue levels of the long-chain (n-3) FA EPA and DHA; SDA in soybean oil appears to be a cost-effective and sustainable plant-based source that could contribute to reaching recommended levels of (n-3) FA intake, but more research and surveillance is needed; and adding SDA-enriched soybean oil to foods should be considered as a natural fortification approach to improving (n-3) FA status in the US and other populations. References for these conclusions and recommendations can be found in the articles included in the supplement.


Subject(s)
Cardiotonic Agents/administration & dosage , Fatty Acids, Omega-3/administration & dosage , Dietary Supplements , Docosahexaenoic Acids/administration & dosage , Eicosapentaenoic Acid/administration & dosage , Evidence-Based Medicine , Humans , Nutrition Policy , Soybean Oil/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...