Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
2.
Int J Mol Sci ; 24(20)2023 Oct 11.
Article in English | MEDLINE | ID: mdl-37894760

ABSTRACT

Diabetic cardiomyopathy is a critical diabetes-mediated co-morbidity characterized by cardiac dysfunction and heart failure, without predisposing hypertensive or atherosclerotic conditions. Metabolic insulin resistance, promoting hyperglycemia and hyperlipidemia, is the primary cause of diabetes-related disorders, but ambiguous tissue-specific insulin sensitivity has shed light on the importance of identifying a unified target paradigm for both the glycemic and non-glycemic context of type 2 diabetes (T2D). Several studies have indicated hyperactivation of the mammalian target of rapamycin (mTOR), specifically complex 1 (mTORC1), as a critical mediator of T2D pathophysiology by promoting insulin resistance, hyperlipidemia, inflammation, vasoconstriction, and stress. Moreover, mTORC1 inhibitors like rapamycin and their analogs have shown significant benefits in diabetes and related cardiac dysfunction. Recently, FDA-approved anti-hyperglycemic sodium-glucose co-transporter 2 inhibitors (SGLT2is) have gained therapeutic popularity for T2D and diabetic cardiomyopathy, even acknowledging the absence of SGLT2 channels in the heart. Recent studies have proposed SGLT2-independent drug mechanisms to ascertain their cardioprotective benefits by regulating sodium homeostasis and mimicking energy deprivation. In this review, we systematically discuss the role of mTORC1 as a unified, eminent target to treat T2D-mediated cardiac dysfunction and scrutinize whether SGLT2is can target mTORC1 signaling to benefit patients with diabetic cardiomyopathy. Further studies are warranted to establish the underlying cardioprotective mechanisms of SGLT2is under diabetic conditions, with selective inhibition of cardiac mTORC1 but the concomitant activation of mTORC2 (mTOR complex 2) signaling.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Cardiomyopathies , Hyperlipidemias , Insulin Resistance , Sodium-Glucose Transporter 2 Inhibitors , Humans , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Diabetic Cardiomyopathies/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Sodium-Glucose Transporter 2 , Mechanistic Target of Rapamycin Complex 2/metabolism , Sodium/metabolism , Hyperlipidemias/drug therapy
3.
Proc Natl Acad Sci U S A ; 120(39): e2302878120, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37722058

ABSTRACT

Although tumor-intrinsic fatty acid ß-oxidation (FAO) is implicated in multiple aspects of tumorigenesis and progression, the impact of this metabolic pathway on cancer cell susceptibility to immunotherapy remains unknown. Here, we report that cytotoxicity of killer T cells induces activation of FAO and upregulation of carnitine palmitoyltransferase 1A (CPT1A), the rate-limiting enzyme of FAO in cancer cells. The repression of CPT1A activity or expression renders cancer cells more susceptible to destruction by cytotoxic T lymphocytes. Our mechanistic studies reveal that FAO deficiency abrogates the prosurvival signaling in cancer cells under immune cytolytic stress. Furthermore, we identify T cell-derived IFN-γ as a major factor responsible for induction of CPT1A and FAO in an AMPK-dependent manner, indicating a dynamic interplay between immune effector cells and tumor targets. While cancer growth in the absence of CPT1A remains largely unaffected, established tumors upon FAO inhibition become significantly more responsive to cellular immunotherapies including chimeric antigen receptor-engineered human T cells. Together, these findings uncover a mode of cancer resistance and immune editing that can facilitate immune escape and limit the benefits of immunotherapies.


Subject(s)
Carnitine O-Palmitoyltransferase , Neoplasms , Humans , Carnitine O-Palmitoyltransferase/genetics , Cytotoxicity, Immunologic , Fatty Acids , Lipid Metabolism , Neoplasms/therapy , T-Lymphocytes, Cytotoxic
4.
J Biol Chem ; 299(7): 104915, 2023 07.
Article in English | MEDLINE | ID: mdl-37315790

ABSTRACT

Biological functions of the highly conserved ubiquitin-like protein 5 (UBL5) are not well understood. In Caenorhabditis elegans, UBL5 is induced under mitochondrial stress to mount the mitochondrial unfolded protein response (UPR). However, the role of UBL5 in the more prevalent endoplasmic reticulum (ER) stress-UPR in the mammalian system is unknown. In the present work, we demonstrated that UBL5 was an ER stress-responsive protein, undergoing rapid depletion in mammalian cells and livers of mice. The ER stress-induced UBL5 depletion was mediated by proteasome-dependent yet ubiquitin-independent proteolysis. Activation of the protein kinase R-like ER kinase arm of the UPR was essential and sufficient for inducing UBL5 degradation. RNA-Seq analysis of UBL5-regulated transcriptome revealed that multiple death pathways were activated in UBL5-silenced cells. In agreement with this, UBL5 knockdown induced severe apoptosis in culture and suppressed tumorigenicity of cancer cells in vivo. Furthermore, overexpression of UBL5 protected specifically against ER stress-induced apoptosis. These results identify UBL5 as a physiologically relevant survival regulator that is proteolytically depleted by the UPR-protein kinase R-like ER kinase pathway, linking ER stress to cell death.


Subject(s)
Cell Death , Endoplasmic Reticulum Stress , Ubiquitins , eIF-2 Kinase , Animals , Mice , Apoptosis , eIF-2 Kinase/metabolism , Ubiquitins/genetics , Ubiquitins/metabolism , Unfolded Protein Response
5.
Nat Commun ; 14(1): 2745, 2023 05 12.
Article in English | MEDLINE | ID: mdl-37173314

ABSTRACT

Proteins of the Hsp110 family are molecular chaperones that play important roles in protein homeostasis in eukaryotes. The pathogenic fungus Candida albicans, which causes infections in humans, has a single Hsp110, termed Msi3. Here, we provide proof-of-principle evidence supporting fungal Hsp110s as targets for the development of new antifungal drugs. We identify a pyrazolo[3,4-b] pyridine derivative, termed HLQ2H (or 2H), that inhibits the biochemical and chaperone activities of Msi3, as well as the growth and viability of C. albicans. Moreover, the fungicidal activity of 2H correlates with its inhibition of in vivo protein folding. We propose 2H and related compounds as promising leads for development of new antifungals and as pharmacological tools for the study of the molecular mechanisms and functions of Hsp110s.


Subject(s)
Antifungal Agents , Candida albicans , Humans , Antifungal Agents/pharmacology , Molecular Chaperones , Protein Folding
6.
Eur J Pharmacol ; 949: 175724, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37059377

ABSTRACT

Berberine is approved for the treatment of intestinal infections and diarrhea and has been shown to have anti-inflammatory and anti-tumor effects in pathological intestinal tissues. However, it is unclear whether the anti-inflammatory effect of berberine contributes to its anti-tumor effect on colitis-associated colorectal cancer (CAC). In this study, we found that berberine effectively inhibited tumorigenesis and protected against colon shortening in CAC mouse model. Immunohistochemistry results showed a reduction in the number of macrophage infiltrations in the colon following berberine treatment. Further analysis revealed that most of the infiltrated macrophages were pro-inflammatory M1 type, which berberine effectively limited. However, in another CRC model without chronic colitis, berberine had no significant effect on tumor number or colon length. In vitro studies demonstrated that berberine treatment significantly reduced the percentage of M1 type and levels of Interleukin-1ß (IL-1ß), Interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). Additionally, miR-155-5p level was down-regulated, and suppressor of cytokine signaling 1 (SOCS1) expression was up-regulated in berberine-treated cells. Notably, the miR-155-5p inhibitor attenuated the regulatory effects of berberine on SOCS1 signaling and macrophage polarization. Altogether, our findings suggest that the inhibitory effect of berberine on CAC development is dependent on its anti-inflammatory activity. Moreover, miR-155-5p may be involved in the pathogenesis of CAC by regulating M1 macrophage polarization, and berberine could be a promising protective agent against miR-155-5p-mediated CAC. This study provides new insights into pharmacologic mechanisms of berberine and supports the possibility that other anti-miR-155-5p drugs may be beneficial in the treatment of CAC.


Subject(s)
Berberine , Colitis-Associated Neoplasms , MicroRNAs , Mice , Animals , Berberine/pharmacology , Berberine/therapeutic use , MicroRNAs/genetics , MicroRNAs/metabolism , Colitis-Associated Neoplasms/drug therapy , Macrophages , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/metabolism , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism
7.
J Pharm Pharmacol ; 74(9): 1353-1363, 2022 Sep 01.
Article in English | MEDLINE | ID: mdl-35641094

ABSTRACT

OBJECTIVES: This study was aimed to explore whether and how berberine suppresses colon cancer cell metastasis via lipid modulation. METHODS: Lipid accumulation was measured by an oil red O staining kit. The expression of proteins and message RNA was detected by Western blot and quantitative real-time PCR. The interaction of sterol-regulatory element-binding proteins cleavage-activating protein (SCAP) with promyelocytic leukaemia zinc finger (PLZF) was confirmed by co-immunoprecipitation assay. Expressions of fatty acid synthase (FASN) and PLZF were knocked down by specific small interfering RNA. KEY FINDINGS: Berberine inhibited the migration and invasion of HCT-8, HCT-116 and HT-29 cells. Moreover, it was observed that berberine decreased lipid droplet accumulation. FASN knockdown abolished the inhibitory effects of berberine on cell migration and invasion. Further investigation revealed that berberine induced the ubiquitination degradation of SCAP. And PLZF interacted with SCAP and promoted its ubiquitination, which was inhibited by berberine treatment. Silence of PLZF impaired the effects of berberine on SCAP ubiquitination and lipogenesis. CONCLUSIONS: Berberine suppressed lipogenesis via promotion of PLZF-mediated SCAP ubiquitination, thereby inhibiting colon cancer cell metastasis.


Subject(s)
Berberine , Colonic Neoplasms , Leukemia , Berberine/pharmacology , Colonic Neoplasms/drug therapy , Humans , Lipids , Lipogenesis , Sterols , Ubiquitination , Zinc Fingers
9.
Methods Mol Biol ; 2455: 49-62, 2022.
Article in English | MEDLINE | ID: mdl-35212985

ABSTRACT

Fatty acid beta oxidation (FAO) is a predominant bioenergetic pathway in mammals. Substantial investigations have demonstrated that FAO activity is dysregulated in many pathophysiological conditions including nonalcoholic steatohepatitis (NASH). Convenient and quantitative assays of FAO activities are important for studies of cell metabolism and the biological relevance of FAO to health and diseases. However, most current FAO assays are based on non-physiological culture conditions, measure FAO activity indirectly or lack adequate quantification. We herein describe details of practical protocols for measurement of basal and genetically or pharmacologically regulated FAO activities in the mammalian system. We also discuss the advantages and disadvantages of these assays in the context of experimental purposes.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Energy Metabolism , Lipolysis , Liver/metabolism , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/metabolism
10.
Protein Sci ; 31(4): 797-810, 2022 04.
Article in English | MEDLINE | ID: mdl-34941000

ABSTRACT

Hsp70s are ubiquitous and highly conserved molecular chaperones. They play crucial roles in maintaining cellular protein homeostasis. It is well established that Hsp70s use the energy of ATP hydrolysis to ADP to power the chaperone activity regardless of the cellular locations and isoforms. Binding immunoglobin protein (BiP), the major member of Hsp70s in the endoplasmic reticulum, is essential for protein folding and quality control. Unexpectedly, our structural analysis of BiP demonstrated a novel ATP hydrolysis to AMP during crystallization under the acidic conditions. Our biochemical studies confirmed this newly discovered ATP to AMP hydrolysis in solutions. Unlike the canonical ATP to ADP hydrolysis observed for Hsp70s, this ATP hydrolysis to AMP depends on the substrate-binding domain of BiP and is inhibited by the binding of a peptide substrate. Intriguingly, this ATP to AMP hydrolysis is unique to BiP, not shared by two representative Hsp70 proteins from the cytosol. Taken together, this novel and unique ATP to AMP hydrolysis may provide a potentially new direction for understanding the activity and cellular function of BiP.


Subject(s)
Carrier Proteins , HSP70 Heat-Shock Proteins , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Carrier Proteins/metabolism , HSP70 Heat-Shock Proteins/chemistry , Humans , Hydrolysis , Protein Binding
11.
FASEB J ; 34(10): 13935-13948, 2020 10.
Article in English | MEDLINE | ID: mdl-32851734

ABSTRACT

Epithelial ovarian carcinoma tissues express high levels of tumor necrosis factor-alpha (TNF-α) and other inflammatory cytokines. The underlying mechanism leading to the abnormal TNF-α expression in ovarian cancer remains poorly understood. In the current study, we demonstrated that lysophosphatidic acid (LPA), a lipid mediator present in ascites of ovarian cancer patients, induced expression of TNF-α mRNA and release of TNF-α protein in ovarian cancer cells. LPA also induced expression of interleukin-1ß (IL-1ß) mRNA but no significant increase in IL-1ß protein was detected. LPA enhanced TNF-α mRNA through NF-κB-mediated transcriptional activation. Inactivation of ADAM17, a disintegrin and metalloproteinase, with a specific inhibitor TMI-1 or by shRNA knockdown prevented ovarian cancer cells from releasing TNF-α protein in response to LPA, indicating that LPA-mediated TNF-α production relies on both transcriptional upregulations of the TNF-α gene and the activity of ADAM17, the membrane-associated TNF-α-converting enzyme. Like many other biological responses to LPA, induction of TNF-α by LPA also depended on the transactivation of the epidermal growth factor receptor (EGFR). Interestingly, our results revealed that ADAM17 was also the shedding protease responsible for the transactivation of EGFR by LPA in ovarian cancer cells. To explore the biological outcomes of LPA-induced TNF-α, we examined the effects of a TNF-α neutralizing antibody and recombinant TNF-α soluble receptor on LPA-stimulated expression of pro-tumorigenic cytokines and chemokines overexpressed in ovarian cancer. Blockade of TNF-α signaling significantly reduced the production of IL-8, IL-6, and CXCL1, suggesting a hierarchy of mechanisms contributing to the robust expression of the inflammatory mediators in response to LPA in ovarian cancer cells. In contrast, TNF-α inhibition did not affect LPA-dependent cell proliferation. Taken together, our results establish that the bioactive lipid LPA drives the expression of TNF-α to regulate an inflammatory network in ovarian cancer.


Subject(s)
Lysophospholipids/pharmacology , Ovarian Neoplasms/metabolism , Tumor Necrosis Factor-alpha/metabolism , ADAM17 Protein/genetics , ADAM17 Protein/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics
12.
Sci Rep ; 10(1): 1450, 2020 01 29.
Article in English | MEDLINE | ID: mdl-31996743

ABSTRACT

Fatty acid oxidation (FAO) is a key bioenergetic pathway often dysregulated in diseases. The current knowledge on FAO regulators in mammalian cells is limited and sometimes controversial. Previous FAO analyses involve nonphysiological culture conditions or lack adequate quantification. We herein described a convenient and quantitative assay to monitor dynamic FAO activities of mammalian cells in physiologically relevant settings. The method enabled us to assess various molecular and pharmacological modulators of the FAO pathway in established cell lines, primary cells and mice. Surprisingly, many previously proposed FAO inhibitors such as ranolazine and trimetazidine lacked FAO-interfering activity. In comparison, etomoxir at low micromolar concentrations was sufficient to saturate its target proteins and to block cellular FAO function. Oxfenicine, on the other hand, acted as a partial inhibitor of FAO. As another class of FAO inhibitors that transcriptionally repress FAO genes, antagonists of peroxisome proliferator-activated receptors (PPARs), particularly that of PPARα, significantly decreased cellular FAO activity. Our assay also had sufficient sensitivity to monitor upregulation of FAO in response to environmental glucose depletion and other energy-demanding cues. Altogether this study provided a reliable FAO assay and a clear picture of biological properties of potential FAO modulators in the mammalian system.


Subject(s)
Fatty Acids/metabolism , Glycine/analogs & derivatives , Mitochondria/metabolism , PPAR alpha/metabolism , Animals , Energy Metabolism , Epoxy Compounds/pharmacology , Female , Glycine/pharmacology , Humans , MCF-7 Cells , Mice , Mice, Inbred C57BL , Oxidation-Reduction , PPAR alpha/antagonists & inhibitors , Ranolazine/pharmacology , Trimetazidine/pharmacology
13.
Cancers (Basel) ; 13(1)2020 Dec 29.
Article in English | MEDLINE | ID: mdl-33383736

ABSTRACT

BACKGROUND: Paclitaxel-induced peripheral neuropathy (PIPN) is a major adverse effect of this chemotherapeutic agent that is used in the treatment of a number of solid malignancies. PIPN leads notably to burning pain, cold and mechanical allodynia. PIPN is thought to be a consequence of alterations of mitochondrial function, hyperexcitability of neurons, nerve fiber loss, oxidative stress and neuroinflammation in dorsal root ganglia (DRG) and spinal cord (SC). Therefore, reducing neuroinflammation could potentially attenuate neuropathy symptoms. Peroxisome proliferator-activated receptor-α (PPAR-α) nuclear receptors that modulate inflammatory responses can be targeted by non-selective agonists, such as fenofibrate, which is used in the treatment of dyslipidemia. METHODS: Our studies tested the efficacy of a fenofibrate diet (0.2% and 0.4%) in preventing the development of PIPN. Paclitaxel (8 mg/kg) was administered via 4 intraperitoneal (i.p.) injections in C57BL/6J mice (both male and female). Mechanical and cold hypersensitivity, wheel running activity, sensory nerve action potential (SNAP), sciatic nerve histology, intra-epidermal fibers, as well as the expression of PPAR-α and neuroinflammation were evaluated in DRG and SC. RESULTS: Fenofibrate in the diet partially prevented the development of mechanical hypersensitivity but completely prevented cold hypersensitivity and the decrease in wheel running activity induced by paclitaxel. The reduction in SNAP amplitude induced by paclitaxel was also prevented by fenofibrate. Our results indicate that suppression of paclitaxel-induced pain by fenofibrate involves the regulation of PPAR-α expression through reduction in neuroinflammation. Finally, co-administration of paclitaxel and the active metabolite of fenofibrate (fenofibric acid) did not interfere with the suppression of tumor cell growth or clonogenicity by paclitaxel in ovarian and breast cancer cell lines. CONCLUSIONS: Taken together, our results show the therapeutic potential of fenofibrate in the prevention of PIPN development.

14.
Biochem Pharmacol ; 174: 113776, 2020 04.
Article in English | MEDLINE | ID: mdl-31874145

ABSTRACT

Lipid metabolism is a significant section of energy homeostasis, and it affects the development of various cancers. Previous studies have revealed that berberine has strong anticancer and blood lipid-lowering effects. Here, we further investigated the effects of berberine on cell proliferation and lipogenesis in colon cancer cells and the relationship between the two effects. We found that berberine inhibited cell proliferation by inducing G0/G1 phase cell cycle arrest in colon cancer cells. Moreover, the expressions of key lipogenic enzymes were down-regulated by berberine and led to the suppressed lipid synthesis, which was linked to cell proliferation via Wnt/ß-catenin pathway. Importantly, berberine inhibited sterol regulatory element-binding protein-1 (SREBP-1) activation and SREBP cleavage-activating protein (SCAP) expression, resulting in the downregulation of these lipogenic enzymes. Knockdown of SCAP by shRNA could abolish the effect of berberine on SREBP-1 activation. Besides the inhibitory effects in vitro, berberine suppressed the growth and lipogenesis of colon cancer xenograft in a SCAP-dependent manner as well. Together, our results suggest that berberine may serve as a candidate against tumor growth of colon cancer partially through targeting SCAP/SREBP-1 pathway driving lipogenesis.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Berberine/pharmacology , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Intracellular Signaling Peptides and Proteins/metabolism , Lipogenesis/drug effects , Membrane Proteins/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Antineoplastic Agents, Phytogenic/therapeutic use , Berberine/therapeutic use , Caco-2 Cells , Cell Line, Tumor , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Humans , Signal Transduction , Xenograft Model Antitumor Assays
15.
Adv Cancer Res ; 143: 195-253, 2019.
Article in English | MEDLINE | ID: mdl-31202359

ABSTRACT

Fundamental metabolic pathways are essential for mammalian cells to provide energy, precursors for biosynthesis of macromolecules, and reducing power for redox regulation. While dysregulated metabolism (e.g., aerobic glycolysis also known as the Warburg effect) has long been recognized as a hallmark of cancer, recent discoveries of metabolic reprogramming in immune cells during their activation and differentiation have led to an emerging concept of "immunometabolism." Considering the recent success of cancer immunotherapy in the treatment of several cancer types, increasing research efforts are being made to elucidate alterations in metabolic profiles of cancer and immune cells during their interplays in the setting of cancer progression and immunotherapy. In this review, we summarize recent advances in studies of metabolic reprogramming in cancer as well as differentiation and functionality of various immune cells. In particular, we will elaborate how distinct metabolic pathways in the tumor microenvironment cause functional impairment of immune cells and contribute to immune evasion by cancer. Lastly, we highlight the potential of metabolically reprogramming the tumor microenvironment to promote effective and long-lasting antitumor immunity for improved immunotherapeutic outcomes.


Subject(s)
Antineoplastic Agents/therapeutic use , Immune System/immunology , Immunotherapy/methods , Neoplasms/drug therapy , Signal Transduction/drug effects , T-Lymphocytes/metabolism , Tumor Microenvironment/immunology , Animals , Cellular Reprogramming , Energy Metabolism , Glycolysis , Humans , Immune System/drug effects , Immune System/metabolism , Metabolome , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes/immunology
16.
J Lipid Res ; 60(5): 972-980, 2019 05.
Article in English | MEDLINE | ID: mdl-30796085

ABSTRACT

The widely expressed lysophosphatidic acid (LPA) selective receptor 4 (LPAR4) contributes to vascular development in mice and zebrafish. LPAR4 regulates endothelial permeability, lymphocyte migration, and hematopoiesis, which could contribute to atherosclerosis. We investigated the role of LPAR4 in experimental atherosclerosis elicited by adeno-associated virus expressing PCSK9 to lower LDL receptor levels. After 20 weeks on a Western diet, cholesterol levels and lipoprotein distribution were similar in WT male and Lpar4Y/- mice (P = 0.94). The atherosclerotic lesion area in the proximal aorta and arch was ∼25% smaller in Lpar4Y/- mice (P = 0.009), and less atherosclerosis was detected in Lpar4Y/- mice at any given plasma cholesterol. Neutral lipid accumulation in aortic root sections occupied ∼40% less area in Lpar4Y/- mice (P = 0.001), and CD68 expression was ∼25% lower (P = 0.045). No difference in α-smooth muscle actin staining was observed. Bone marrow-derived macrophages isolated from Lpar4Y/- mice displayed significantly increased upregulation of the M2 marker Arg1 in response to LPA compared with WT cells. In aortic root sections from Lpar4Y/- mice, heightened M2 "repair" macrophage marker expression was detected by CD206 staining (P = 0.03). These results suggest that LPAR4 may regulate the recruitment of specific sets of macrophages or their phenotypic switching in a manner that could influence the development of atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Receptors, Purinergic/deficiency , Receptors, Purinergic/metabolism , Animals , Cells, Cultured , Lysophospholipids/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
17.
Cancer Lett ; 435: 92-100, 2018 10 28.
Article in English | MEDLINE | ID: mdl-30102953

ABSTRACT

Cancer cells undergo metabolic reprogramming such as enhanced aerobic glycolysis, mutations in the tricarboxylic acid cycle enzymes, and upregulation of de novo lipid synthesis and glutaminolysis. These alterations are pivotal to the development and maintenance of the malignant phenotype of cancer cells in unfavorable tumor microenvironment or metastatic sites. Although mitochondrial fatty acid ß-oxidation (FAO) is a primary bioenergetic source, it has not been generally recognized as part of the metabolic landscape of cancer. The last few years, however, have seen a dramatic change in the view of cancer relevance of the FAO pathway. Many recent studies have provided significant evidence to support a "lipolytic phenotype" of cancer. FAO, like other well-defined metabolic pathways involved in cancer, is dysregulated in diverse human malignancies. Cancer cells rely on FAO for proliferation, survival, stemness, drug resistance, and metastatic progression. FAO is also reprogrammed in cancer-associated immune and other host cells, which may contribute to immune suppression and tumor-promoting microenvironment. This article reviews and puts into context our current understanding of multi-faceted roles of FAO in oncogenesis as well as anti-cancer therapeutic opportunities posed by the FAO pathway.


Subject(s)
Energy Metabolism , Fatty Acids/metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Cell Transformation, Neoplastic/metabolism , Humans , Lipid Metabolism , Lipolysis , Neoplasms/pathology , Oxidation-Reduction , Tumor Microenvironment
18.
Int Immunopharmacol ; 57: 121-131, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29482156

ABSTRACT

Berberine has been reported to have protective effects in colitis treatment. However, the detailed mechanisms remain unclear. Herein, we demonstrated that berberine could protect against dextran sulfate sodium (DSS)-induced colitis in mice by regulating macrophage polarization. In the colitis mouse model, berberine ameliorated DSS-induced colon shortening and colon tissue injury. Moreover, berberine-treated mice showed significant reduction in the disease activity index (DAI), pro-inflammatory cytokines expression and macrophages infiltration compared with the DSS-treated mice. Notably, berberine significantly reduced the percentage of M1 macrophages. In vitro analysis also confirmed the inhibitory effects of berberine on macrophages M1 polarization in RAW267.4 cells. Further investigation showed that berberine promoted AKT1 expression in mRNA and protein level. Silence of AKT1 abolished the inhibitory effect of berberine on macrophages M1 polarization. The berberine-induced AKT1 expression promoted suppressers of cytokine signaling (SOCS1) activation, which inhibited nuclear factor-kappa B (NF-κB) phosphorylation. In addition, we also found that berberine activated AKT1/SOCS1 signaling pathway but inhibited p65 phosphorylation in macrophages in vivo. Therefore, we concluded that berberine played a regulatory role in macrophages M1 polarization in DSS-induced colitis via AKT1/SOCS1/NF-κB signaling pathway. This unexpected property of berberine may provide a potential explanation for its protective effects in colitis treatment.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Berberine/therapeutic use , Cell Differentiation/drug effects , Colitis/drug therapy , Macrophages/physiology , Animals , Colitis/chemically induced , Cytokines/metabolism , Dextran Sulfate , Disease Models, Animal , Humans , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RAW 264.7 Cells , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism , Th1 Cells/immunology
19.
Cancers (Basel) ; 11(1)2018 Dec 31.
Article in English | MEDLINE | ID: mdl-30602670

ABSTRACT

The recent progresses in understanding of cancer glycolytic phenotype have offered new strategies to manage ovarian cancer and other malignancies. However, therapeutic targeting of glycolysis to treat cancer remains unsuccessful due to complex mechanisms of tumor glycolysis and the lack of selective, potent and safe glycolytic inhibitors. Recently, BAY-876 was identified as a new-generation inhibitor of glucose transporter 1 (GLUT1), a GLUT isoform commonly overexpressed but functionally poorly defined in ovarian cancer. Notably, BAY-876 has not been evaluated in any cell or preclinical animal models since its discovery. We herein took advantage of BAY-876 and molecular approaches to study GLUT1 regulation, targetability, and functional relevance to cancer glycolysis. The anti-tumor activity of BAY-876 was evaluated with ovarian cancer cell line- and patient-derived xenograft (PDX) models. Our results show that inhibition of GLUT1 is sufficient to block basal and stress-regulated glycolysis, and anchorage-dependent and independent growth of ovarian cancer cells. BAY-876 dramatically inhibits tumorigenicity of both cell line-derived xenografts and PDXs. These studies provide direct evidence that GLUT1 is causally linked to the glycolytic phenotype in ovarian cancer. BAY-876 is a potent blocker of GLUT1 activity, glycolytic metabolism and ovarian cancer growth, holding promise as a novel glycolysis-targeted anti-cancer agent.

20.
J Pharmacol Exp Ther ; 364(1): 110-119, 2018 01.
Article in English | MEDLINE | ID: mdl-29042416

ABSTRACT

Chemotherapy-induced peripheral neuropathy (CIPN), a consequence of peripheral nerve fiber dysfunction or degeneration, continues to be a dose-limiting and debilitating side effect during and/or after cancer chemotherapy. Paclitaxel, a taxane commonly used to treat breast, lung, and ovarian cancers, causes CIPN in 59-78% of cancer patients. Novel interventions are needed due to the current lack of effective CIPN treatments. Our studies were designed to investigate whether nicotine can prevent and/or reverse paclitaxel-induced peripheral neuropathy in a mouse model of CIPN, while ensuring that nicotine will not stimulate lung tumor cell proliferation or interfere with the antitumor properties of paclitaxel. Male C57BL/6J mice received paclitaxel every other day for a total of four injections (8 mg/kg, i.p.). Acute (0.3-0.9 mg/kg, i.p.) and chronic (24 mg/kg per day, s.c.) administration of nicotine respectively reversed and prevented paclitaxel-induced mechanical allodynia. Blockade of the antinociceptive effect of nicotine with mecamylamine and methyllycaconitine suggests that the reversal of paclitaxel-induced mechanical allodynia is primarily mediated by the α7 nicotinic acetylcholine receptor subtype. Chronic nicotine treatment also prevented paclitaxel-induced intraepidermal nerve fiber loss. Notably, nicotine neither promoted proliferation of A549 and H460 non-small cell lung cancer cells nor interfered with paclitaxel-induced antitumor effects, including apoptosis. Most importantly, chronic nicotine administration did not enhance Lewis lung carcinoma tumor growth in C57BL/6J mice. These data suggest that the nicotinic acetylcholine receptor-mediated pathways may be promising drug targets for the prevention and treatment of CIPN.


Subject(s)
Hyperalgesia/drug therapy , Hyperalgesia/prevention & control , Nicotine/pharmacology , Paclitaxel/adverse effects , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Bridged-Ring Compounds/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Disease Models, Animal , Hyperalgesia/chemically induced , Lung Neoplasms/drug therapy , Male , Mice , Mice, Inbred C57BL , Paclitaxel/pharmacology , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/prevention & control , Receptors, Cholinergic/metabolism , Taxoids/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...