Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Mol Oncol ; 17(5): 839-856, 2023 05.
Article in English | MEDLINE | ID: mdl-35838343

ABSTRACT

Small-cell lung cancer (SCLC) is an aggressive neuroendocrine subtype of lung cancer with poor patient prognosis. However, the mechanisms that regulate SCLC progression and metastasis remain undefined. Here, we show that the expression of the slit guidance ligand 2 (SLIT2) tumor suppressor gene is reduced in SCLC tumors relative to adjacent normal tissue. In addition, the expression of the SLIT2 receptor, roundabout guidance receptor 1 (ROBO1), is upregulated. We find a positive association between SLIT2 expression and the Yes1 associated transcriptional regulator (YAP1)-expressing SCLC subtype (SCLC-Y), which shows a better prognosis. Using genetically engineered SCLC cells, adenovirus gene therapy, and preclinical xenograft models, we show that SLIT2 overexpression or the deletion of ROBO1 restricts tumor growth in vitro and in vivo. Mechanistic studies revealed significant inhibition of myeloid-derived suppressor cells (MDSCs) and M2-like tumor-associated macrophages (TAMs) in the SCLC tumors. In addition, SLIT2 enhances M1-like and phagocytic macrophages. Molecular analysis showed that ROBO1 knockout or SLIT2 overexpression suppresses the transforming growth factor beta 1 (TGF-ß1)/ß-catenin signaling pathway in both tumor cells and macrophages. Overall, we find that SLIT2 and ROBO1 have contrasting effects on SCLC tumors. SLIT2 suppresses, whereas ROBO1 promotes, SCLC growth by regulating the Tgf-ß1/glycogen synthase kinase-3 beta (GSK3)/ß-catenin signaling pathway in tumor cells and TAMs. These studies indicate that SLIT2 could be used as a novel therapeutic agent against aggressive SCLC.


Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Transforming Growth Factor beta1/pharmacology , beta Catenin/metabolism , Nerve Tissue Proteins/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3/pharmacology , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Signal Transduction , Small Cell Lung Carcinoma/genetics , Lung Neoplasms/genetics , Macrophages/metabolism
2.
Cancers (Basel) ; 14(5)2022 Feb 24.
Article in English | MEDLINE | ID: mdl-35267489

ABSTRACT

Chemotherapy forms the backbone of current treatments for many patients with advanced non-small-cell lung cancer (NSCLC). However, the survival rate is low in these patients due to the development of drug resistance, including cisplatin resistance. In this study, we developed a novel strategy to combat the growth of cisplatin-resistant (CR) NSCLC cells. We have shown that treatment with the plant-derived, non-psychotropic small molecular weight molecule, cannabidiol (CBD), significantly induced apoptosis of CR NSCLC cells. In addition, CBD treatment significantly reduced tumor progression and metastasis in a mouse xenograft model and suppressed cancer stem cell properties. Further mechanistic studies demonstrated the ability of CBD to inhibit the growth of CR cell lines by reducing NRF-2 and enhancing the generation of reactive oxygen species (ROS). Moreover, we show that CBD acts through Transient Receptor Potential Vanilloid-2 (TRPV2) to induce apoptosis, where TRPV2 is expressed on human lung adenocarcinoma tumors. High expression of TRPV2 correlates with better overall survival of lung cancer patients. Our findings identify CBD as a novel therapeutic agent targeting TRPV2 to inhibit the growth and metastasis of this aggressive cisplatin-resistant phenotype in NSCLC.

3.
J Exp Clin Cancer Res ; 41(1): 54, 2022 Feb 08.
Article in English | MEDLINE | ID: mdl-35135586

ABSTRACT

BACKGROUND: Molecular mechanisms underlying inflammation-associated breast tumor growth are poorly studied. S100A7, a pro-inflammatory molecule has been shown to enhance breast cancer growth and metastasis. However, the S100A7-mediated molecular mechanisms in enhancing tumor growth and metastasis are unclear. METHODS: Human breast cancer tissue and plasma samples were used to analyze the expression of S100A7, cPLA2, and PGE2. S100A7-overexpressing or downregulated human metastatic breast cancer cells were used to evaluate the S100A7-mediated downstream signaling mechanisms. Bi-transgenic mS100a7a15 overexpression, TNBC C3 (1)/Tag transgenic, and humanized patient-derived xenograft mouse models and cPLA2 inhibitor (AACOCF3) were used to investigate the role of S100A7/cPLA2/PGE2 signaling in tumor growth and metastasis. Additionally, CODEX, a highly advanced multiplexed imaging was employed to delineate the effects of S100A7/cPLA2 inhibition on the recruitment of various immune cells. RESULTS: In this study, we found that S100A7 and cPLA2 are highly expressed and correlate with decreased overall survival in breast cancer patients. Further mechanistic studies revealed that S100A7/RAGE signaling promotes the expression of cPLA2 to mediate its oncogenic effects. Pharmacological inhibition of cPLA2 suppressed S100A7-mediated tumor growth and metastasis in multiple pre-clinical models including transgenic and humanized patient-derived xenograft (PDX) mouse models. The attenuation of cPLA2 signaling reduced S100A7-mediated recruitment of immune-suppressive myeloid cells in the tumor microenvironment (TME). Interestingly, we discovered that the S100A7/cPLA2 axis enhances the immunosuppressive microenvironment by increasing prostaglandin E2 (PGE2). Furthermore, CO-Detection by indEXing (CODEX) imaging-based analyses revealed that cPLA2 inhibition increased the infiltration of activated and proliferating CD4+ and CD8+ T cells in the TME. In addition, CD163+ tumor associated-macrophages were positively associated with S100A7 and cPLA2 expression in malignant breast cancer patients. CONCLUSIONS: Our study provides new mechanistic insights on the cross-talk between S100A7/cPLA2 in enhancing breast tumor growth and metastasis by generating an immunosuppressive TME that inhibits the infiltration of cytotoxic T cells. Furthermore, our studies indicate that S100A7/cPLA2 could be used as novel prognostic marker and cPLA2 inhibitors as promising drugs against S100A7-overexpressing aggressive breast cancer.


Subject(s)
Breast Neoplasms/genetics , Phospholipases A2, Cytosolic/antagonists & inhibitors , S100 Calcium Binding Protein A7/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Female , Humans , Mice , Tumor Microenvironment
4.
Front Immunol ; 12: 753477, 2021.
Article in English | MEDLINE | ID: mdl-34777365

ABSTRACT

Slit2 exerts antitumor effects in various cancers; however, the underlying mechanism, especially its role in regulating the immune, especially in the bone marrow niche, system is still unknown. Elucidating the behavior of macrophages in tumor progression can potentially improve immunotherapy. Using a spontaneous mammary tumor virus promoter-polyoma middle T antigen (PyMT) breast cancer mouse model, we observed that Slit2 increased the abundance of antitumor M1 macrophage in the bone marrow upon differentiation in vitro. Moreover, myeloablated PyMT mice injected with Slit2-treated bone marrow allografts showed a marked reduction in tumor growth, with enhanced recruitment of M1 macrophage in their tumor stroma. Mechanistic studies revealed that Slit2 significantly enhanced glycolysis and reduced fatty acid oxidation in bone marrow-derived macrophages (BMDMs). Slit2 treatment also altered mitochondrial respiration metabolites in macrophages isolated from healthy human blood that were treated with plasma from breast cancer patients. Overall, this study, for the first time, shows that Slit2 increases BMDM polarization toward antitumor phenotype by modulating immune-metabolism. Furthermore, this study provides evidence that soluble Slit2 could be developed as novel therapeutic strategy to enhance antitumor immune response.


Subject(s)
Intercellular Signaling Peptides and Proteins/physiology , Macrophage Activation/drug effects , Macrophages/drug effects , Mammary Neoplasms, Experimental/therapy , Metabolome/drug effects , Nerve Tissue Proteins/physiology , Adult , Aged , Animals , Antigens, Polyomavirus Transforming/genetics , Culture Media, Conditioned , Female , Glycolysis/drug effects , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Lipopolysaccharide Receptors/analysis , Macrophages/immunology , Macrophages/metabolism , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Transgenic , Middle Aged , Monocytes/drug effects , Monocytes/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Radiation Chimera , TOR Serine-Threonine Kinases/physiology , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/chemistry , Tumor Burden
5.
Bioelectricity ; 3(1): 92-100, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-34476380

ABSTRACT

Background: Induced electric fields (iEFs) control directional breast cancer cell migration. While the connection between migration and metabolism is appreciated in the context of cancer and metastasis, effects of iEFs on metabolic pathways especially as they relate to migration, remain unexplored. Materials and Methods: Quantitative cell migration data in the presence and absence of an epidermal growth factor (EGF) gradient in the microfluidic bidirectional microtrack assay was retrospectively analyzed for additional effects of iEFs on cell motility and directionality. Surrogate markers of oxidative phosphorylation (succinate dehydrogenase [SDH] activity) and glycolysis (lactate dehydrogenase activity) were assessed in MDA-MB-231 breast cancer cells and normal MCF10A mammary epithelial cells exposed to iEFs and EGF. Results: Retrospective analysis of migration results suggests that iEFs increase forward cell migration speeds while extending the time cells spend migrating slowly in the reverse direction or remaining stationary. Furthermore, in the presence of EGF, iEFs differentially altered flux through oxidative phosphorylation in MDA-MB-231 cells and glycolysis in MCF10A cells. Conclusions: iEFs interfere with MDA-MB-231 cell migration, potentially, by altering mitochondrial metabolism, observed as an inhibition of SDH activity in the presence of EGF. The energy intensive process of migration in these highly metastatic breast cancer cells may be hindered by iEFs, thus, through hampering of oxidative phosphorylation.

6.
Cancer Lett ; 509: 115-120, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33798632

ABSTRACT

The growing burden of obesity and incidence of the aggressive triple negative breast cancer (TNBC) is a challenge, especially amongst vulnerable populations with unmet medical needs and higher mortality from breast cancer. While some mechanisms linking obesity and TNBC have been identified, the complex nature of pathogenesis, in both obesity as well as TNBC poses a real challenge in establishing a causative role of obesity in risk of TNBC. In this review article, we discuss pathological mechanisms identified in the tumor microenvironment (TME) as well as the obese microenvironment (OME), such as inflammation, insulin resistance and survival pathways that contribute to the development and progression of TNBC. Insights into the cross-talk between TME and OME, and their contribution to TNBC development and progression, may pave the way for personalized therapies against TNBC progression, relapse and metastasis.


Subject(s)
Adipose Tissue/metabolism , Obesity/metabolism , Triple Negative Breast Neoplasms/metabolism , Tumor Microenvironment , Adipose Tissue/pathology , Animals , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Disease Progression , Energy Metabolism , Female , Humans , Incidence , Inflammation Mediators/metabolism , Insulin Resistance , Obesity/epidemiology , Obesity/pathology , Obesity/therapy , Risk Factors , Signal Transduction , Triple Negative Breast Neoplasms/epidemiology , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/therapy
7.
Cell Death Dis ; 11(9): 774, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32943608

ABSTRACT

Triple-negative breast cancer (TNBC), defined as loss of estrogen, progesterone, and Her2 receptors, is a subtype of highly aggressive breast cancer with worse prognosis and poor survival rate. Macrophage migration inhibitory factor (MIF) is a pleiotropic pro-inflammatory cytokine aberrantly expressed in many solid tumors and known to promote tumor progression and metastasis. However, its role in TNBC progression and metastasis is unexplored. Here we have shown that in TNBC patients, MIF expression was significantly enriched in the tumor compared to adjacent normal tissue. Using publically available patient datasets, we showed that MIF overexpression correlates with worse survival in TNBC compared to other hormonal status. Orthotopic implantation of TNBC cells into MIF knockout mice showed reduced tumor growth compared to wild-type mice. In addition, we have shown that MIF downregulation inhibits TNBC growth and progression in a syngeneic mouse model. We further showed that CPSI-1306, a small-molecule MIF inhibitor, inhibits the growth of TNBC cells in vitro. Mechanistic studies revealed that CPSI-1306 induces intrinsic apoptosis by alteration in mitochondrial membrane potential, cytochrome c (Cyt c) release, and activation of different caspases. In addition, CPSI-1306 inhibits the activation of cell survival and proliferation-related molecules. CPSI-1306 treatment also reduced the tumor growth and metastasis in orthotopic mouse models of mammary carcinoma. CPSI-1306 treatment of tumor-bearing mice significantly inhibited TNBC growth and pulmonary metastasis in a dose-dependent manner. Histological analysis of xenograft tumors revealed a higher number of apoptotic cells in CPSI-1306-treated tumors compared to vehicle controls. Our studies, for the first time, show that MIF overexpression in TNBC enhances growth and metastasis. Taken together, our results indicate that using small molecular weight MIF inhibitors could be a promising strategy to inhibit TNBC progression and metastasis.


Subject(s)
Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Triple Negative Breast Neoplasms/drug therapy , Animals , Apoptosis , Caspases/metabolism , Cell Movement , Cell Survival , Cytochromes c/metabolism , Disease Progression , Enzyme Activation , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , In Vitro Techniques , Inflammation , Intramolecular Oxidoreductases/antagonists & inhibitors , Isoxazoles/pharmacology , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Morpholines/pharmacology , Neoplasm Metastasis , Neoplasm Transplantation , Treatment Outcome , Triple Negative Breast Neoplasms/pathology , Wound Healing
8.
J Biol Chem ; 295(4): 969-980, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31822562

ABSTRACT

The rising prevalence of type 1 diabetes (T1D) over the past decades has been linked to lifestyle changes, but the underlying mechanisms are largely unknown. Recent findings point to gut-associated mechanisms in the control of T1D pathogenesis. In nonobese diabetic (NOD) mice, a model of T1D, diabetes development accelerates after deletion of the Toll-like receptor 4 (TLR4). We hypothesized that altered intestinal functions contribute to metabolic alterations, which favor accelerated diabetes development in TLR4-deficient (TLR4-/-) NOD mice. In 70-90-day-old normoglycemic (prediabetic) female NOD TLR4+/+ and NOD TLR4-/- mice, gut morphology and microbiome composition were analyzed. Parameters of lipid metabolism, glucose homeostasis, and mitochondrial respiratory activity were measured in vivo and ex vivo Compared with NOD TLR4+/+ mice, NOD TLR4-/- animals showed lower muscle mass of the small intestine, higher abundance of Bacteroidetes, and lower Firmicutes in the large intestine, along with lower levels of circulating short-chain fatty acids (SCFA). These changes are associated with higher body weight, hyperlipidemia, and severe insulin and glucose intolerance, all occurring before the onset of diabetes. These mice also exhibited insulin resistance-related abnormalities of energy metabolism, such as lower total respiratory exchange rates and higher hepatic oxidative capacity. Distinct alterations of gut morphology and microbiota composition associated with reduction of circulating SCFA may contribute to metabolic disorders promoting the progression of insulin-deficient diabetes/T1D development.


Subject(s)
Diabetes Mellitus, Experimental/microbiology , Diabetes Mellitus, Experimental/pathology , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/pathology , Animals , Blood Glucose/metabolism , Body Weight , Diabetes Mellitus, Experimental/blood , Energy Metabolism , Fatty Acids/metabolism , Homeostasis , Lipopolysaccharides/metabolism , Liver/pathology , Mice, Inbred C57BL , Mice, Inbred NOD , Models, Biological , Oxidation-Reduction , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/metabolism , alpha-2-HS-Glycoprotein/metabolism
9.
Commun Biol ; 2: 303, 2019.
Article in English | MEDLINE | ID: mdl-31428691

ABSTRACT

Interactions between cells and their environment influence key physiologic processes such as their propensity to migrate. However, directed migration controlled by extrinsically applied electrical signals is poorly understood. Using a novel microfluidic platform, we found that metastatic breast cancer cells sense and respond to the net direction of weak (∼100 µV cm-1), asymmetric, non-contact induced Electric Fields (iEFs). iEFs inhibited EGFR (Epidermal Growth Factor Receptor) activation, prevented formation of actin-rich filopodia, and hindered the motility of EGF-treated breast cancer cells. The directional effects of iEFs were nullified by inhibition of Akt phosphorylation. Moreover, iEFs in combination with Akt inhibitor reduced EGF-promoted motility below the level of untreated controls. These results represent a step towards isolating the coupling mechanism between cell motility and iEFs, provide valuable insights into how iEFs target multiple diverging cancer cell signaling mechanisms, and demonstrate that electrical signals are a fundamental regulator of cancer cell migration.


Subject(s)
Breast Neoplasms/pathology , Cell Movement , Electromagnetic Fields , Actins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Down-Regulation/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Female , Humans , Neoplasm Metastasis , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
10.
Adv Exp Med Biol ; 1162: 51-61, 2019.
Article in English | MEDLINE | ID: mdl-31332734

ABSTRACT

The family of chemical structures that interact with a cannabinoid receptor are broadly termed cannabinoids. Traditionally known for their psychotropic effects and their use as palliative medicine in cancer, cannabinoids are very versatile and are known to interact with several orphan receptors besides cannabinoid receptors (CBR) in the body. Recent studies have shown that several key pathways involved in cell growth, differentiation and, even metabolism and apoptosis crosstalk with cannabinoid signaling. Several of these pathways including AKT, EGFR, and mTOR are known to contribute to tumor development and metastasis, and cannabinoids may reverse their effects, thereby by inducing apoptosis, autophagy and modulating the immune system. In this book chapter, we explore how cannabinoids regulate diverse signaling mechanisms in cancer and immune cells within the tumor microenvironment and whether they impart a therapeutic effect. We also provide some important insight into the role of cannabinoids in cellular and whole body metabolism in the context of tumor inhibition. Finally, we highlight recent and ongoing clinical trials that include cannabinoids as a therapeutic strategy and several combinational approaches towards novel therapeutic opportunities in several invasive cancer conditions.


Subject(s)
Cannabinoids/pharmacology , Neoplasms , Receptors, Cannabinoid/physiology , Signal Transduction , Apoptosis , Humans , Neoplasm Metastasis , Tumor Microenvironment
11.
Oncogene ; 37(32): 4428-4442, 2018 08.
Article in English | MEDLINE | ID: mdl-29720724

ABSTRACT

The chemokine CXCL12 has been shown to regulate breast tumor growth, however, its mechanism in initiating distant metastasis is not well understood. Here, we generated a novel conditional allele of Cxcl12 in mice and used a fibroblast-specific Cre transgene along with various mammary tumor models to evaluate CXCL12 function in the breast cancer metastasis. Ablation of CXCL12 in stromal fibroblasts of mice significantly delayed the time to tumor onset and inhibited distant metastasis in different mouse models. Elucidation of mechanisms using in vitro and in vivo model systems revealed that CXCL12 enhances tumor cell intravasation by increasing vascular permeability and expansion of a leaky tumor vasculature. Furthermore, our studies revealed CXCL12 enhances permeability by recruiting endothelial precursor cells and decreasing endothelial tight junction and adherence junction proteins. High expression of stromal CXCL12 in large cohort of breast cancer patients was directly correlated to blood vessel density and inversely correlated to recurrence and overall patient survival. In addition, our analysis revealed that stromal CXCL12 levels in combination with number of CD31+ blood vessels confers poorer patient survival compared to individual protein level. However, no correlation was observed between epithelial CXCL12 and patient survival or blood vessel density. Our findings describe the novel interactions between fibroblasts-derived CXCL12 and endothelial cells in facilitating tumor cell intrvasation, leading to distant metastasis. Overall, our studies indicate that cross-talk between fibroblast-derived CXCL12 and endothelial cells could be used as novel biomarker and strategy for developing tumor microenvironment based therapies against aggressive and metastatic breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chemokine CXCL12/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Neoplasm Invasiveness/pathology , Animals , Cell Line , Cell Line, Tumor , Cell Movement/physiology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Mammary Neoplasms, Animal , Mice , Mice, Transgenic , Neoplasm Metastasis/pathology , Tumor Microenvironment/physiology
12.
Exp Mol Med ; 49(11): e394, 2017 11 17.
Article in English | MEDLINE | ID: mdl-29147011

ABSTRACT

Increased oxidative stress is implicated in the pathogenesis of experimental diabetic neuropathy, but translational evidence in recent-onset diabetes is scarce. We aimed to determine whether markers of systemic oxidative stress are associated with diabetic sensorimotor polyneuropathy (DSPN) in recent-onset diabetes. In this cross-sectional study, we measured serum concentrations of extracellular superoxide dismutase (SOD3), thiobarbituric acid reactive substances (TBARS), and reduced glutathione (GSH) in 107 type 1 and 215 type 2 diabetes patients from the German Diabetes Study baseline cohort and 37 glucose-tolerant individuals (controls). DSPN was defined by electrophysiological and clinical criteria (Toronto Consensus, 2011). SOD3 and GSH concentrations were lower in individuals with type 1 and type 2 diabetes compared with concentrations in controls (P<0.0001). In contrast, the TBARS concentration was higher in participants with type 1 diabetes and type 2 diabetes compared with levels in controls (P<0.0001). In addition, the SOD3 concentration was higher in participants with type 1 diabetes compared to concentrations in those with type 2 diabetes (P<0.0001). A low SOD3 concentration was associated with DSPN in individuals with type 1 diabetes (ß=-0.306, P=0.002), type 2 diabetes (ß=-0.164, P=0.017), and in both groups combined (ß=-0.206, P=0.0003). Lower SOD3 concentrations were associated with decreased motor nerve conduction velocity (NCV) in men and, to a lesser degree, with reduced sensory NCV in women with diabetes. In conclusion, several biomarkers of oxidative stress are altered in recent-onset diabetes, with only a lower SOD3 concentration being linked to the presence of DSPN, suggesting a role for reduced extracellular antioxidative defense against superoxide in the early development of DSPN.


Subject(s)
Diabetes Complications , Polyneuropathies/blood , Polyneuropathies/etiology , Superoxide Dismutase/blood , Adult , Biomarkers , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 2/complications , Female , Humans , Male , Oxidative Stress , Peripheral Nerves/physiopathology , Polyneuropathies/diagnosis , Sex Factors , Symptom Assessment
13.
Diabetes ; 66(8): 2241-2253, 2017 08.
Article in English | MEDLINE | ID: mdl-28490610

ABSTRACT

Nonalcoholic fatty liver disease is associated with hepatic insulin resistance and may result primarily from increased hepatic de novo lipogenesis (PRIM) or secondarily from adipose tissue lipolysis (SEC). We studied mice with hepatocyte- or adipocyte-specific SREBP-1c overexpression as models of PRIM and SEC. PRIM mice featured increased lipogenic gene expression in the liver and adipose tissue. Their selective, liver-specific insulin resistance was associated with increased C18:1-diacylglycerol content and protein kinase Cε translocation. SEC mice had decreased lipogenesis mediated by hepatic cholesterol responsive element-binding protein and featured portal/lobular inflammation along with total, whole-body insulin resistance. Hepatic mitochondrial respiration transiently increased and declined with aging along with higher muscle reactive oxygen species production. In conclusion, hepatic insulin resistance originates from lipotoxicity but not from lower mitochondrial capacity, which can even transiently adapt to increased peripheral lipolysis. Peripheral insulin resistance is prevented during increased hepatic lipogenesis only if adipose tissue lipid storage capacity is preserved.


Subject(s)
Insulin Resistance/physiology , Lipogenesis/genetics , Non-alcoholic Fatty Liver Disease/physiopathology , Sterol Regulatory Element Binding Protein 1/metabolism , Adipocytes/metabolism , Animals , Diglycerides/metabolism , Female , Hepatocytes/metabolism , Lipolysis/genetics , Liver/cytology , Liver/physiopathology , Mice , Mice, Transgenic , Non-alcoholic Fatty Liver Disease/genetics , Protein Kinase C-epsilon/metabolism , Protein Transport , Reactive Oxygen Species/metabolism
14.
Diabetes ; 65(7): 1849-57, 2016 07.
Article in English | MEDLINE | ID: mdl-27207512

ABSTRACT

Type 1 diabetes has been recently linked to nonalcoholic fatty liver disease (NAFLD), which is known to associate with insulin resistance, obesity, and type 2 diabetes. However, the role of insulin resistance and hyperglycemia for hepatic energy metabolism is yet unclear. To analyze early abnormalities in hepatic energy metabolism, we examined 55 patients with recently diagnosed type 1 diabetes. They underwent hyperinsulinemic-normoglycemic clamps with [6,6-(2)H2]glucose to assess whole-body and hepatic insulin sensitivity. Hepatic γATP, inorganic phosphate (Pi), and triglyceride concentrations (hepatocellular lipid content [HCL]) were measured with multinuclei magnetic resonance spectroscopy ((31)P/(1)H-MRS). Glucose-tolerant humans served as control (CON) (n = 57). Whole-body insulin sensitivity was 44% lower in patients than in age- and BMI-matched CON. Hepatic γATP was 15% reduced (2.3 ± 0.6 vs. 2.7 ± 0.6 mmol/L, P < 0.001), whereas hepatic Pi and HCL were similar in patients when compared with CON. Across all participants, hepatic γATP correlated negatively with glycemia and oxidized LDL. Carriers of the PPARG G allele (rs1801282) and noncarriers of PPARGC1A A allele (rs8192678) had 21 and 13% lower hepatic ATP concentrations. Variations in genes controlling oxidative metabolism contribute to a reduction in hepatic ATP in the absence of NAFLD, suggesting that alterations in hepatic mitochondrial function may precede diabetes-related liver diseases.


Subject(s)
Adenosine Triphosphate/metabolism , Adipose Tissue/metabolism , Diabetes Mellitus, Type 1/genetics , Energy Metabolism/genetics , Liver/metabolism , Adult , Alleles , Body Mass Index , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Female , Glucose Clamp Technique , Humans , Insulin Resistance/genetics , Lipid Metabolism/genetics , Liver/pathology , Male , Oxidative Stress/physiology , PPAR gamma/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Phosphates/metabolism , Triglycerides/metabolism
15.
Metabolism ; 64(12): 1629-39, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26455399

ABSTRACT

For long the presence of insulin resistance in type 1 diabetes has been questioned. Detailed metabolic analyses revealed 12%-61% and up to 20% lower whole-body (skeletal muscle) and hepatic insulin sensitivity in type 1 diabetes, depending on the population studied. Type 1 diabetes patients feature impaired muscle adenosine triphosphate (ATP) synthesis and enhanced oxidative stress, predominantly relating to hyperglycemia. They may also exhibit abnormal fasting and postprandial glycogen metabolism in liver, while the role of hepatic energy metabolism for insulin resistance remains uncertain. Recent rodent studies point to tissue-specific differences in the mechanisms underlying insulin resistance. In non-obese diabetic mice, increased lipid availability contributes to muscle insulin resistance via diacylglycerol/protein kinase C isoforms. Furthermore, humans with type 1 diabetes respond to lifestyle modifications or metformin by 20%-60% increased whole-body insulin sensitivity, likely through improvement in both glycemic control and oxidative phosphorylation. Intensive insulin treatment and islet transplantation also increase but fail to completely restore whole-body and hepatic insulin sensitivity. In conclusion, insulin resistance is a feature of type 1 diabetes, but more controlled trials are needed to address its contribution to disease progression, which might help to optimize treatment and reduce comorbidities.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Insulin Resistance , Adipose Tissue/metabolism , Animals , Humans , Liver/metabolism , Mice , Oxidative Stress
16.
Cell Metab ; 21(5): 739-46, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25955209

ABSTRACT

The association of hepatic mitochondrial function with insulin resistance and non-alcoholic fatty liver (NAFL) or steatohepatitis (NASH) remains unclear. This study applied high-resolution respirometry to directly quantify mitochondrial respiration in liver biopsies of obese insulin-resistant humans without (n = 18) or with (n = 16) histologically proven NAFL or with NASH (n = 7) compared to lean individuals (n = 12). Despite similar mitochondrial content, obese humans with or without NAFL had 4.3- to 5.0-fold higher maximal respiration rates in isolated mitochondria than lean persons. NASH patients featured higher mitochondrial mass, but 31%-40% lower maximal respiration, which associated with greater hepatic insulin resistance, mitochondrial uncoupling, and leaking activity. In NASH, augmented hepatic oxidative stress (H2O2, lipid peroxides) and oxidative DNA damage (8-OH-deoxyguanosine) was paralleled by reduced anti-oxidant defense capacity and increased inflammatory response. These data suggest adaptation of the liver ("hepatic mitochondrial flexibility") at early stages of obesity-related insulin resistance, which is subsequently lost in NASH.


Subject(s)
Fatty Liver/pathology , Liver/pathology , Mitochondria, Liver/pathology , Non-alcoholic Fatty Liver Disease/pathology , Adult , Cell Respiration , Fatty Liver/complications , Fatty Liver/metabolism , Female , Humans , Hydrogen Peroxide/metabolism , Insulin Resistance , Lipid Peroxidation , Liver/metabolism , Male , Middle Aged , Mitochondria, Liver/metabolism , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/complications , Obesity/metabolism , Oxidative Stress
17.
Diabetes ; 63(11): 3856-67, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24917575

ABSTRACT

Although insulin resistance is known to underlie type 2 diabetes, its role in the development of type 1 diabetes has been gaining increasing interest. In a model of type 1 diabetes, the nonobese diabetic (NOD) mouse, we found that insulin resistance driven by lipid- and glucose-independent mechanisms is already present in the liver of prediabetic mice. Hepatic insulin resistance is associated with a transient rise in mitochondrial respiration followed by increased production of lipid peroxides and c-Jun N-terminal kinase activity. At the onset of diabetes, increased adipose tissue lipolysis promotes myocellular diacylglycerol accumulation. This is paralleled by increased myocellular protein kinase C θ activity and serum fetuin A levels. Muscle mitochondrial oxidative capacity is unchanged at the onset but decreases at later stages of diabetes. In conclusion, hepatic and muscle insulin resistance manifest at different stages and involve distinct cellular mechanisms during the development of diabetes in the NOD mouse.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin Resistance/physiology , Animals , Diabetes Mellitus, Type 2/blood , Disease Models, Animal , JNK Mitogen-Activated Protein Kinases/metabolism , Lipid Peroxides/metabolism , Liver/metabolism , Mice , Prediabetic State/metabolism , Protein Kinase C/metabolism , alpha-2-HS-Glycoprotein/metabolism
18.
Diabetes Care ; 37(2): 468-74, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24026561

ABSTRACT

OBJECTIVE: Muscle insulin resistance has been implicated in the development of steatosis and dyslipidemia by changing the partitioning of postprandial substrate fluxes. Also, insulin resistance may be due to reduced mitochondrial function. We examined the association between mitochondrial activity, insulin sensitivity, and steatosis in a larger human population. RESEARCH DESIGN AND METHODS: We analyzed muscle mitochondrial activity from ATP synthase flux (fATP) and ectopic lipids by multinuclei magnetic resonance spectroscopy from 113 volunteers with and without diabetes. Insulin sensitivity was assessed from M values using euglycemic-hyperinsulinemic clamps and/or from oral glucose insulin sensitivity (OGIS) using oral glucose tolerance tests. RESULTS: Muscle fATP correlated negatively with hepatic lipid content and HbA1c. After model adjustment for study effects and other confounders, fATP showed a strong negative correlation with hepatic lipid content and a positive correlation with insulin sensitivity and fasting C-peptide. The negative correlation of muscle fATP with age, HbA1c, and plasma free fatty acids was weakened after adjustment. Body mass, muscle lipid contents, plasma lipoproteins, and triglycerides did not associate with fATP. CONCLUSIONS: The association of impaired muscle mitochondrial activity with hepatic steatosis supports the concept of a close link between altered muscle and liver energy metabolism as early abnormalities promoting insulin resistance.


Subject(s)
Fasting , Fatty Liver/metabolism , Mitochondria, Muscle/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Adult , Female , Humans , Insulin Resistance , Magnetic Resonance Spectroscopy , Male , Middle Aged , Mitochondria, Muscle/enzymology , Young Adult
19.
Curr Diabetes Rev ; 9(2): 146-60, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23094754

ABSTRACT

Diabetic retinopathy (DR) is the leading cause of blindness amongst the working-age population, and diabetes accelerated cardiovascular disease (CVD) the commonest cause of death in diabetic patients. Although, there is evidence suggesting a close association between DR and CVD, particularly in patients with Type 2 diabetes, the pathophysiology underlying the link is unclear. Here we review common risk factors and pathogenic mechanisms linking DR and CVD, and aim to highlight the need for a more holistic view of the management of diabetes and its complications. The understanding of the link between the two complications could eventually lead to refined management strategies and improved patient outcomes in the expanding diabetes epidemic.


Subject(s)
Atherosclerosis/physiopathology , Blindness/physiopathology , Diabetic Angiopathies/physiopathology , Diabetic Retinopathy/physiopathology , Glycation End Products, Advanced/metabolism , Hexosamines/metabolism , Protein Kinase C/metabolism , Animals , Apoptosis , Atherosclerosis/metabolism , Atherosclerosis/mortality , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/mortality , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/mortality , Disease Progression , Humans , Leukostasis , Mice , Mice, Inbred NOD , Mice, Knockout , Oxidative Stress , Risk Factors , Signal Transduction
20.
ISRN Ophthalmol ; 2013: 343560, 2013.
Article in English | MEDLINE | ID: mdl-24563789

ABSTRACT

Diabetes is now regarded as an epidemic, with the population of patients expected to rise to 380 million by 2025. Tragically, this will lead to approximately 4 million people around the world losing their sight from diabetic retinopathy, the leading cause of blindness in patients aged 20 to 74 years. The risk of development and progression of diabetic retinopathy is closely associated with the type and duration of diabetes, blood glucose, blood pressure, and possibly lipids. Although landmark cross-sectional studies have confirmed the strong relationship between chronic hyperglycaemia and the development and progression of diabetic retinopathy, the underlying mechanism of how hyperglycaemia causes retinal microvascular damage remains unclear. Continued research worldwide has focussed on understanding the pathogenic mechanisms with the ultimate goal to prevent DR. The aim of this paper is to introduce the multiple interconnecting biochemical pathways that have been proposed and tested as key contributors in the development of DR, namely, increased polyol pathway, activation of protein kinase C (PKC), increased expression of growth factors such as vascular endothelial growth factor (VEGF) and insulin-like growth factor-1 (IGF-1), haemodynamic changes, accelerated formation of advanced glycation endproducts (AGEs), oxidative stress, activation of the renin-angiotensin-aldosterone system (RAAS), and subclinical inflammation and capillary occlusion. New pharmacological therapies based on some of these underlying pathogenic mechanisms are also discussed.

SELECTION OF CITATIONS
SEARCH DETAIL
...