Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article En | MEDLINE | ID: mdl-35210363

Cancer-associated cachexia (CAC) is a hypermetabolic syndrome characterized by unintended weight loss due to the atrophy of adipose tissue and skeletal muscle. A phenotypic switch from white to beige adipocytes, a phenomenon called browning, accelerates CAC by increasing the dissipation of energy as heat. Addressing the mechanisms of white adipose tissue (WAT) browning in CAC, we now show that cachexigenic tumors activate type 2 immunity in cachectic WAT, generating a neuroprotective environment that increases peripheral sympathetic activity. Increased sympathetic activation, in turn, results in increased neuronal catecholamine synthesis and secretion, ß-adrenergic activation of adipocytes, and induction of WAT browning. Two genetic mouse models validated this progression of events. 1) Interleukin-4 receptor deficiency impeded the alternative activation of macrophages, reduced sympathetic activity, and restrained WAT browning, and 2) reduced catecholamine synthesis in peripheral dopamine ß-hydroxylase (DBH)-deficient mice prevented cancer-induced WAT browning and adipose atrophy. Targeting the intraadipose macrophage-sympathetic neuron cross-talk represents a promising therapeutic approach to ameliorate cachexia in cancer patients.


Adipose Tissue, Brown/pathology , Cachexia/pathology , Cell Communication , Neoplasms/complications , Neurons/pathology , Sympathetic Nervous System/pathology , Animals , Cachexia/etiology , Cachexia/metabolism , Gene Expression , Heterografts , Humans , Mice , Neoplasms/metabolism , Receptors, Adrenergic, beta/metabolism , Thermogenesis
2.
Nephrol Dial Transplant ; 36(9): 1577-1584, 2021 08 27.
Article En | MEDLINE | ID: mdl-32542315

Chronic kidney disease (CKD) is a progressive multisystem condition with yet undefined mechanistic drivers and multiple implicated soluble factors. If identified, these factors could be targeted for therapeutic intervention for a disease that currently lacks specific treatment. There is increasing preclinical evidence that the heparin/endothelial glycocalyx-binding molecule midkine (MK) has a pathological role in multiple CKD-related, organ-specific disease processes, including CKD progression, hypertension, vascular and cardiac disease, bone disease and CKD-related cancers. Concurrent with this are studies documenting increases in circulating and urine MK proportional to glomerular filtration rate (GFR) loss in CKD patients and evidence that administering soluble MK reverses the protective effects of MK deficiency in experimental kidney disease. This review summarizes the growing body of evidence supporting MK's potential role in driving CKD-related multisystem disease, including MK's relationship with the endothelial glycocalyx, the deranged MK levels and glycocalyx profile in CKD patients and a proposed model of MK organ interplay in CKD disease processes and highlights the importance of ongoing research into MK's potential as a therapeutic target.


Multiple Organ Failure , Renal Insufficiency, Chronic , Glomerular Filtration Rate , Humans , Longitudinal Studies , Midkine , Renal Insufficiency, Chronic/etiology
3.
Sci Rep ; 10(1): 14499, 2020 09 02.
Article En | MEDLINE | ID: mdl-32879333

Midkine (MDK), a heparin-binding growth factor cytokine, is involved in the pathogenesis of kidney diseases by augmenting leukocyte trafficking and activation. Animal models and small case control studies have implicated MDK as a pathological biomarker in chronic kidney diseases (CKD), however this is yet to be confirmed in prospective human studies. In a prospective study of 499 elderly, predominantly Caucasian women aged over 70 years the association between serum MDK collected in 1998, and renal function change and the risk of CKD-related hospitalisations and deaths at 5 and 14.5 years, respectively, was examined. Baseline serum MDK was not associated with 5-year change in estimated glomerular filtration rate using the CKD Epidemiology Collaboration creatinine and cystatin C equation (Standardised ß = - 0.09, 95% confidence interval - 3.76-0.48, p = 0.129), 5-year rapid decline in renal function (odds ratio = 0.97, 95% confidence interval 0.46-2.02, p = 0.927) or the risk of 14.5-year CKD-related hospitalisations and deaths (hazard ratio = 1.27, 95% confidence interval .66-2.46, p = 0.470) before or after adjusting for major risk factors. In conclusion, in this cohort of elderly women with normal or mildly impaired renal function, serum MDK was not associated with renal function change or future CKD-related hospitalisations and deaths, suggesting that MDK may not be an early biomarker for progression of CKD.


Glomerular Filtration Rate , Kidney Failure, Chronic/therapy , Midkine/blood , Aged , Aging , Biomarkers/metabolism , Disease Progression , Female , Humans , Longitudinal Studies , Multivariate Analysis , Prognosis , Proportional Hazards Models , Prospective Studies , Risk Factors , Western Australia
4.
Nutrients ; 10(5)2018 May 22.
Article En | MEDLINE | ID: mdl-29786640

Vascular calcification (VC) as a manifestation of perturbed mineral balance, is associated with aging, diabetes and kidney dysfunction, as well as poorer patient outcomes. Due to the current limited understanding of the pathophysiology of vascular calcification, the development of effective preventative and therapeutic strategies remains a significant clinical challenge. Recent evidence suggests that traditional risk factors for cardiovascular disease, such as left ventricular hypertrophy and dyslipidaemia, fail to account for clinical observations of vascular calcification. Therefore, more complex underlying processes involving physiochemical changes to mineral balance, vascular remodelling and perturbed hormonal responses such as parathyroid hormone (PTH) and fibroblast growth factor 23 (FGF-23) are likely to contribute to VC. In particular, VC resulting from modifications to calcium, phosphate and vitamin D homeostasis has been recently elucidated. Notably, deregulation of vitamin D metabolism, dietary calcium intake and renal mineral handling are associated with imbalances in systemic calcium and phosphate levels and endothelial cell dysfunction, which can modulate both bone and soft tissue calcification. This review addresses the current understanding of VC pathophysiology, with a focus on the pathogenic role of vitamin D that has provided new insights into the mechanisms of VC.


Blood Vessels/metabolism , Calcium, Dietary/metabolism , Osteogenesis , Vascular Calcification/metabolism , Vitamin D/metabolism , Animals , Blood Vessels/pathology , Blood Vessels/physiopathology , Calcium, Dietary/adverse effects , Fibroblast Growth Factor-23 , Homeostasis , Humans , Risk Factors , Signal Transduction , Vascular Calcification/diagnosis , Vascular Calcification/epidemiology , Vascular Calcification/physiopathology , Vitamin D/adverse effects , Vitamin D Deficiency/epidemiology , Vitamin D Deficiency/metabolism , Vitamin D Deficiency/physiopathology
5.
Semin Cell Dev Biol ; 54: 68-81, 2016 06.
Article En | MEDLINE | ID: mdl-26529279

Although muscle wasting is the obvious manifestation of cancer cachexia that impacts on patient quality of life, the loss of lipid reserves and metabolic imbalance in adipose tissue also contribute to the devastating impact of cachexia. Depletion of fat depots in cancer patients is more pronounced than loss of muscle and often precedes, or even occurs in the absence of, reduced lean body mass. Rapid mobilisation of triglycerides stored within adipocytes to supply the body with fatty acids in periods of high-energy demand is normally mediated through a well-defined process of lipolysis involving the lipases ATGL, HSL and MGL. Studies into how these lipases contribute to fat loss in cancer cachexia have revealed the prominent role for ATGL in initiating lipolysis during adipose tissue atrophy, together with links between tumour-derived factors and the signalling pathways that control lipid flux within fat cells. The recent findings of increased thermogenesis in brown fat during cancer cachexia indicate that metabolically active adipose tissue contributes to the imbalance in energy homeostasis involved in catabolic wasting. Such energetically futile use of fatty acids liberated from adipose tissue to generate heat represents a maladaptive response in conjunction with anorexia experienced by cancer patients. As IL-6 release by tumours provokes lipolysis and activates the thermogenic programme in brown fat, this review explores the overlap in dysregulated metabolic processes due to inflammatory mediators in cancer cachexia and other disease states characterised by elevated cytokines such as obesity and diabetes.


Adipose Tissue/metabolism , Cachexia/etiology , Cachexia/metabolism , Lipolysis , Neoplasms/complications , Thermogenesis , Animals , Cachexia/physiopathology , Energy Metabolism , Humans , Neoplasms/physiopathology
6.
Pharm Res ; 30(9): 2270-8, 2013 Sep.
Article En | MEDLINE | ID: mdl-23604979

PURPOSE: In many cancer patients, the malignancy causes reduced hepatic drug clearance leading to potentially serious complications from the use of anticancer drugs. The mechanisms underlying this phenomenon are poorly understood. We aimed to identify tumor-associated inflammatory pathways that alter drug response and enhance chemotherapy-associated toxicity. METHODS: We studied inflammatory pathways involved in extra-hepatic tumor mediated repression of CYP3A, a major hepatic drug metabolizing cytochrome P450 subfamily, using a murine Engelbreth-Holm-Swarm sarcoma model. Studies in IL-6 knockout mice determined the source of elevated IL-6 in tumor-bearing animals and monoclonal antibodies against IL-6 were used to intervene in this inflammatory pathway. RESULTS: Our studies confirm elevated plasma IL-6 levels and reveal activation of Jak/Stat and Mapk signalling pathways and acute phase proteins in livers of tumor-bearing mice. Circulating IL-6 was predominantly produced by the tumor xenograft, rather than being host derived. Anti IL-6 antibody intervention partially reversed tumor-mediated inflammation and Cyp3a gene repression. CONCLUSIONS: IL-6 is an important player in cancer-related repression of CYP3A-mediated drug metabolism and activation of the acute phase response. Targeting IL-6 in cancer patients may prove an effective approach to alleviating cancer-related phenomena, such as adverse drug-related outcomes commonly associated with cancer chemotherapy.


Antineoplastic Agents/metabolism , Cytochrome P-450 CYP3A/metabolism , Interleukin-6/immunology , Liver/metabolism , Sarcoma, Experimental/immunology , Animals , Cytochrome P-450 CYP3A/immunology , Interleukin-6/genetics , Liver/drug effects , Liver/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Sarcoma, Experimental/drug therapy , Sarcoma, Experimental/metabolism
8.
Proc Natl Acad Sci U S A ; 109(7): 2302-7, 2012 Feb 14.
Article En | MEDLINE | ID: mdl-22308320

Src tyrosine kinase has long been implicated in colon cancer but much remains to be learned about its substrates. The nuclear receptor hepatocyte nuclear factor 4α (HNF4α) has just recently been implicated in colon cancer but its role is poorly defined. Here we show that c-Src phosphorylates human HNF4α on three tyrosines in an interdependent and isoform-specific fashion. The initial phosphorylation site is a Tyr residue (Y14) present in the N-terminal A/B domain of P1- but not P2-driven HNF4α. Phospho-Y14 interacts with the Src SH2 domain, leading to the phosphorylation of two additional tyrosines in the ligand binding domain (LBD) in P1-HNF4α. Phosphomimetic mutants in the LBD decrease P1-HNF4α protein stability, nuclear localization and transactivation function. Immunohistochemical analysis of approximately 450 human colon cancer specimens (Stage III) reveals that P1-HNF4α is either lost or localized in the cytoplasm in approximately 80% of tumors, and that staining for active Src correlates with those events in a subset of samples. Finally, three SNPs in the human HNF4α protein, two of which are in the HNF4α F domain that interacts with the Src SH3 domain, increase phosphorylation by Src and decrease HNF4α protein stability and function, suggesting that individuals with those variants may be more susceptible to Src-mediated effects. This newly identified interaction between Src kinase and HNF4α has important implications for colon and other cancers.


Cell Nucleus/metabolism , Colonic Neoplasms/enzymology , Hepatocyte Nuclear Factor 4/metabolism , Protein Isoforms/metabolism , src-Family Kinases/metabolism , Cell Line , Colonic Neoplasms/pathology , Hepatocyte Nuclear Factor 4/genetics , Humans , Molecular Mimicry , Phosphorylation , Polymorphism, Single Nucleotide , Protein Isoforms/genetics
9.
Aging (Albany NY) ; 4(2): 133-43, 2012 Feb.
Article En | MEDLINE | ID: mdl-22361433

Cancer cachexia is a highly debilitating paraneoplastic disease observed in more than 50% of patients with advanced cancers and directly contributes to 20% of cancer deaths. Loss of skeletal muscle is a defining characteristic of patients with cancer cachexia and is associated with poor survival. The present study reveals the involvement of a myogenic transcription factor Myocyte Enhancer Factor (MEF) 2C in cancer-induced skeletal muscle wasting. Increased skeletal muscle mRNA expression of Suppressor of Cytokine Signaling (Socs) 3 and the IL-6 receptor indicative of active IL-6 signaling was seen in skeletal muscle of mice bearing the Colon 26 (C26) carcinoma. Loss of skeletal muscle structural integrity and distorted mitochondria were also observed using electron microscopy. Gene and protein expression of MEF2C was significantly downregulated in skeletal muscle from C26-bearing mice. MEF2C gene targets myozenin and myoglobin as well as myokinase were also altered during cachexia, suggesting dysregulated oxygen transport capacity and ATP regeneration in addition to distorted structural integrity. In addition, reduced expression of calcineurin was observed which suggested a potential pathway of MEF2C dysregulation. Together, these effects may limit sarcomeric contractile ability and also predispose skeletal muscle to structural instability; associated with muscle wasting and fatigue in cachexia.


Cachexia/etiology , Cachexia/metabolism , Muscle, Skeletal/metabolism , Myogenic Regulatory Factors/metabolism , Animals , Cachexia/pathology , Calcineurin/metabolism , Cell Line, Tumor , Down-Regulation , Interleukin-6/metabolism , MEF2 Transcription Factors , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Microscopy, Electron, Transmission , Mitochondria, Muscle/pathology , Models, Neurological , Muscle, Skeletal/pathology , Myogenic Regulatory Factors/genetics , Neoplasms, Experimental/complications , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sarcomeres/pathology , Signal Transduction
10.
Methods Mol Biol ; 781: 311-36, 2011.
Article En | MEDLINE | ID: mdl-21877288

Molecular expression patterns have often been used for patient classification in oncology in an effort to improve prognostic prediction and treatment compatibility. This effort is, however, hampered by the highly heterogeneous data often seen in the molecular analysis of cancer. The lack of overall similarity between expression profiles makes it difficult to partition data using conventional data mining tools. In this chapter, the authors introduce a bioinformatics protocol that uses REACTOME pathways and patient-protein network structure (also called topology) as the basis for patient classification.


Computational Biology/methods , Neoplasms/diagnosis , Neoplasms/metabolism , Biomarkers, Tumor/metabolism , Cluster Analysis , Humans , Neoplasms/therapy , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Software , Tandem Mass Spectrometry
11.
Mol Biosyst ; 7(11): 2997-3005, 2011 Nov.
Article En | MEDLINE | ID: mdl-21808808

Quantitative mass spectrometry using iTRAQ was used to identify differentially expressed proteins from 16 colorectal cancer (CRC) tumours compared to patient-paired adjacent normal mucosa. Over 1400 proteins were identified and quantitated, with 118 determined as differentially expressed by >1.3-fold, with false discovery rate < 0.05. Gene Ontology analysis indicated that proteins with increased expression levels in CRC tumours include those associated with glycolysis, calcium binding, and protease inhibition. Proteins with reduced levels in CRC tumours were associated with loss of ATP production through: (i) reduced ß-oxidation of fatty acids, (ii) reduced NADH production by the tricarboxylic acid cycle and (iii) decreased oxidative phosphorylation activity. Additionally, biosyntheses of glycosaminoglycans and proteoglycans were significantly reduced in tumour samples. Validation experiments using immunoblotting and immunohistochemistry (IHC) showed strong concordance with iTRAQ data suggesting that this workflow is suitable for identifying biomarker candidates. We discuss the uses and challenges of this approach to generate biomarker leads for patient prognostication.


Biomarkers, Tumor/analysis , Colorectal Neoplasms/metabolism , Intestinal Mucosa/metabolism , Mass Spectrometry/methods , Aged , Aged, 80 and over , Colorectal Neoplasms/chemistry , Colorectal Neoplasms/pathology , Female , Humans , Immunohistochemistry , Intestinal Mucosa/chemistry , Intestinal Mucosa/pathology , Male , Middle Aged , Proteomics
12.
Clin Cancer Res ; 17(10): 3170-80, 2011 May 15.
Article En | MEDLINE | ID: mdl-21498392

PURPOSE: To determine the mechanisms by which tumors situated in extrahepatic sites can cause profound changes in hepatic drug clearance, contributing to altered drug response and chemotherapy resistance. EXPERIMENTAL DESIGN: We studied in wild-type or transgenic CYP3A4 reporter mice implanted with the murine Engelbreth-Holm-Swarm sarcoma changes in nuclear receptor and hepatic transcription factor expression and/or function, particularly related to CYP3A gene regulation. RESULTS: Repression of hepatic CYP3A induction was dramatic and associated with reduced levels of C/EBPß isoforms, impaired pregnane X receptor, and constitutive androstane receptor function. Unexpectedly, extrahepatic tumors strongly reduced nuclear accumulation of retinoid X receptor alpha (RXRα) in hepatocytes, providing a potential explanation for impaired function of nuclear receptors that rely on RXRα dimerization. Profiling revealed 38 nuclear receptors were expressed in liver with 14 showing between 1.5- and four-fold reduction in expression in livers of tumor-bearing animals, including Car, Trß, Lxrß, Pparα, Errα/ß, Reverbα/ß, and Shp. Altered Pparα and γ induction of target genes provided additional evidence of perturbed hepatic metabolic control elicited by extrahepatic tumors. CONCLUSIONS: Extrahepatic malignancy can affect hepatic drug metabolism by nuclear receptor relocalization and decreased receptor expression and function. These findings could aid the design of intervention strategies to normalize drug clearance and metabolic pathways in cancer patients at risk of chemotherapy-induced toxicity or cancer cachexia.


Liver/metabolism , Neoplasms/metabolism , Pharmaceutical Preparations/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Cell Line, Tumor , Cytochrome P-450 CYP3A/genetics , Down-Regulation/physiology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Inactivation, Metabolic/genetics , Lac Operon , Male , Mice , Mice, Transgenic , Neoplasms/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Sarcoma, Experimental/genetics , Sarcoma, Experimental/pathology
13.
Mol Cell Proteomics ; 10(9): M900538MCP200, 2011 Sep.
Article En | MEDLINE | ID: mdl-20167946

Cancer is well known to be associated with alterations in membrane protein glycosylation (Bird, N. C., Mangnall, D., and Majeed, A. W. (2006) Biology of colorectal liver metastases: A review. J. Surg. Oncol. 94, 68-80; Dimitroff, C. J., Pera, P., Dall'Olio, F., Matta, K. L., Chandrasekaran, E. V., Lau, J. T., and Bernacki, R. J. (1999) Cell surface n-acetylneuraminic acid alpha2,3-galactoside-dependent intercellular adhesion of human colon cancer cells. Biochem. Biophys. Res. Commun. 256, 631-636; and Arcinas, A., Yen, T. Y., Kebebew, E., and Macher, B. A. (2009) Cell surface and secreted protein profiles of human thyroid cancer cell lines reveal distinct glycoprotein patterns. J. Proteome Res. 8, 3958-3968). Equally, it has been well established that tumor-associated inflammation through the release of pro-inflammatory cytokines is a common cause of reduced hepatic drug metabolism and increased toxicity in advanced cancer patients being treated with cytotoxic chemotherapies. However, little is known about the impact of bearing a tumor (and downstream effects like inflammation) on liver membrane protein glycosylation. In this study, proteomic and glycomic analyses were used in combination to determine whether liver membrane protein glycosylation was affected in mice bearing the Engelbreth-Holm Swarm sarcoma. Peptide IPG-IEF and label-free quantitation determined that many enzymes involved in the protein glycosylation pathway specifically; mannosidases (Man1a-I, Man1b-I and Man2a-I), mannoside N-acetylglucosaminyltransferases (Mgat-I and Mgat-II), galactosyltransferases (B3GalT-VII, B4GalT-I, B4GalT-III, C1GalT-I, C1GalT-II, and GalNT-I), and sialyltransferases (ST3Gal-I, ST6Gal-I, and ST6GalNAc-VI) were up-regulated in all livers of tumor-bearing mice (n = 3) compared with nontumor bearing controls (n = 3). In addition, many cell surface lectins: Sialoadhesin-1 (Siglec-1), C-type lectin family 4f (Kupffer cell receptor), and Galactose-binding lectin 9 (Galectin-9) were determined to be up-regulated in the liver of tumor-bearing compared with control mice. Global glycan analysis identified seven N-glycans and two O-glycans that had changed on the liver membrane proteins derived from tumor-bearing mice. Interestingly, α (2,3) sialic acid was found to be up-regulated on the liver membrane of tumor-bearing mice, which reflected the increased expression of its associated sialyltransferase and lectin receptor (siglec-1). The overall increased sialylation on the liver membrane of Engelbreth-Holm Swarm bearing mice correlates with the increased expression of their associated glycosyltransferases and suggests that glycosylation of proteins in the liver plays a role in tumor-induced liver inflammation.


Galactosyltransferases/metabolism , Glycomics/methods , Inflammation/metabolism , Liver/metabolism , Mannosidases/metabolism , Neoplasms/metabolism , Proteome/metabolism , Proteomics/methods , Sarcoma, Experimental/metabolism , Sialyltransferases/metabolism , Animals , Cell Membrane/genetics , Cell Membrane/metabolism , Galactosyltransferases/genetics , Gene Expression Regulation, Neoplastic , Glycosylation , Inflammation/etiology , Inflammation/genetics , Lectins/genetics , Lectins/metabolism , Liver/cytology , Male , Mannosidases/genetics , Mice , Mice, Transgenic , N-Acetylneuraminic Acid/genetics , N-Acetylneuraminic Acid/metabolism , Neoplasm Transplantation , Neoplasms/complications , Neoplasms/genetics , Polysaccharides/genetics , Polysaccharides/metabolism , Proteome/genetics , Receptors, Mitogen/genetics , Receptors, Mitogen/metabolism , Sarcoma, Experimental/complications , Sarcoma, Experimental/genetics , Sialyltransferases/genetics
14.
Int J Biochem Cell Biol ; 42(5): 672-82, 2010 May.
Article En | MEDLINE | ID: mdl-20060928

The orphan nuclear receptor pregnane X receptor regulates enzymes and transport proteins involved in the detoxification and clearance of numerous endobiotic and xenobiotic compounds, including pharmaceutical agents. Multiple alternatively spliced pregnane X receptor isoforms have been identified which are significantly expressed in humans and mice (up to 30% of the total pregnane X receptor transcript), however, little is known about their biological action. We explored functional differences between the major mouse pregnane X receptor isoforms mPXR(431) and mPXR(Delta171-211) that lacks 41 amino acids adjacent to the ligand-binding pocket. Transient transfection assays showed that mPXR(Delta171-211) reduced the basal transcription of cytochrome P450 3A4 and the drug transporter P-glycoprotein/Multi Drug Resistance Protein 1 and directly repressed the regulatory effects of mPXR(431) on these genes. Replacement of the mPXR(Delta171-211) DNA-binding domain with that of GAL4 showed mPXR(Delta171-211) retained its repressive role independent of binding to PXR responsive elements located within the cytochrome P450 3A4 and Multi Drug Resistance Protein 1 regulatory regions. Use of the histone deacetylase inhibitor, trichostatin A, demonstrated that the repressive function of mPXR(Delta171-211) acts independently of histone acetylation state. Protein interaction assays revealed mPXR(Delta171-211) and mPXR(431) differentially bind the obligatory heterodimer partner retinoid X receptor. Furthermore, mPXR(431) and mPXR(Delta171-211) proteins could heterodimerize. These studies demonstrate that the variant mouse PXR isoform, mPXR(Delta171-211), has a distinct repressive function from mPXR(431) in regulating genes encoding important drug metabolizing enzymes and transport proteins.


ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Alternative Splicing , Cytochrome P-450 CYP3A/metabolism , Down-Regulation , Receptors, Steroid/genetics , Receptors, Steroid/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Amino Acid Sequence , Animals , Cell Line , Chlorocebus aethiops , Cytochrome P-450 CYP3A/genetics , DNA-Binding Proteins , Histone Deacetylase Inhibitors/pharmacology , Humans , Male , Mice , Organ Specificity , Pregnane X Receptor , Protein Interaction Domains and Motifs , Protein Isoforms/genetics , Protein Isoforms/physiology , Protein Multimerization , Receptors, Steroid/chemistry , Recombinant Fusion Proteins , Regulatory Elements, Transcriptional/genetics , Retinoid X Receptors/metabolism , Sequence Alignment , Sequence Deletion
15.
Nucl Recept Signal ; 7: e004, 2009 May 08.
Article En | MEDLINE | ID: mdl-19471583

Subcellular compartmentalisation and the intracellular movement of nuclear receptors are major regulatory steps in executing their transcriptional function. Though significant progress has been made in understanding these regulatory processes in cultured mammalian cells, such results have rarely been confirmed within cells of a living mammal. This article describes a simple, time-efficient approach to study the nuclear versus cytoplasmic accumulation of nuclear receptors and the regions of nuclear receptor proteins that govern subcellular trafficking within hepatocytes of live mice. Pregnane X receptor, a xenobiotic-activated member of the nuclear receptor family, was used to exemplify the approach. Using dual-labeled wild-type and mutant PXR expression constructs, we outline their in vivo delivery, simultaneous cellular expression, visualization and categorical classification within hepatocytes of live mice. Using this approach, we identified three mutants that had an altered subcellular distribution in the presence and absence of a PXR ligand. This novel in vivo method complements the current cell culture-based experimental systems in protein subcellular localisation studies.


Cell Culture Techniques/methods , Gene Expression Profiling/methods , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/ultrastructure , Subcellular Fractions/metabolism , Subcellular Fractions/ultrastructure , Cells, Cultured
16.
Expert Opin Drug Metab Toxicol ; 4(2): 137-49, 2008 Feb.
Article En | MEDLINE | ID: mdl-18248309

BACKGROUND: The inability to accurately predict treatment outcomes for cancer patients in terms of tumour response and anticancer drug toxicity is a severe limitation inherent in current approaches to chemotherapy. Many anticancer drugs are metabolically cleared by cytochrome P450 3A4 (CYP3A4), the predominant CYP expressed in liver. CYP3A4 expression exhibits marked interindividual variation and is repressed in acute inflammatory states. OBJECTIVES: (1) To review the relevance of CYP3A4 variability to drug metabolism in the setting of cancer and to understand how inflammation associated with malignancy contributes to both this variability and to adverse treatment outcomes. (2) To examine the relationship between tumour-induced inflammation and repression of CYP3A4 and to explore methods of dosing of anticancer drugs in the setting of advanced cancer. METHODS: Review of relevant literature covering both human and animal studies as well as in vitro mechanistic studies. RESULTS/CONCLUSIONS: Interindividual variability in CYP3A4 expression is a major confounding factor for effective cancer treatment and methods to predict CYP3A4-mediated drug clearance may have clinical utility in this setting. Although acute inflammation has long been recognised to repress drug metabolism, it is now becoming apparent that cancer patients exhibiting clinical and laboratory features of an inflammatory response have reduced expression of CYP3A4 and possibly other genes relevant to anticancer drug disposition.


Antineoplastic Agents/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Neoplasms/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Cytochrome P-450 CYP3A , Gene Expression Regulation, Enzymologic , Humans , Inflammation/etiology , Inflammation/physiopathology , Neoplasm Staging , Neoplasms/physiopathology , Pharmacogenetics
17.
Drug Metab Dispos ; 36(2): 205-16, 2008 Feb.
Article En | MEDLINE | ID: mdl-18218849

This article is a report on a symposium sponsored by the American Society for Pharmacology and Experimental Therapeutics and held at the Experimental Biology 07 meeting in Washington, DC. The presentations discussed the phenomenology, clinical consequences, and underlying mechanisms of cytochrome P450 and drug transporter regulation by inflammatory and infectious stimuli. Although considerable insights into the links between inflammatory mediators and altered hepatic drug clearance pathways have been gained from previous studies with acute inflammatory stimuli, this symposium highlighted recent advances in understanding how these processes operate in other organs and chronic inflammatory states relevant to human diseases. The development of mouse models of live bacterial infection provides excellent opportunities to explore the impact of infection on drug metabolism beyond the well characterized effects of bacterial endotoxin. Altered levels of cytochromes P450 and especially drug transporters due to inflammation in brain, intestine, and placenta have significant implications for the use of many drugs in diverse clinical settings. The consequences of inflammatory cytokine production by tumors for drug safety and efficacy in cancer patients were outlined. Repression of drug clearance pathways by tumor-derived cytokines may result in extreme toxicity to chemotherapy, compromising treatment of many cancers. It is fitting that, in honoring the career contributions and achievements of Dr. Kenneth W. Renton, this symposium reinforced the clinical relevance of this field.


Cytochrome P-450 Enzyme System/metabolism , Infections/metabolism , Inflammation/metabolism , Membrane Transport Proteins/metabolism , Neoplasms/metabolism , Animals , Cytochrome P-450 Enzyme System/genetics , Humans , Pharmaceutical Preparations/metabolism , Transcription, Genetic
18.
Int J Biochem Cell Biol ; 39(3): 478-83, 2007.
Article En | MEDLINE | ID: mdl-17188925

The Pregnane X Receptor (PXR) is pivotal for the body's response to toxic xenobiotics and endogenous metabolites. By acting as a ligand-activated transcription factor, PXR regulates all stages of xenobiotic metabolism and transport and is responsible for important inductive drug interactions. Screening assays to assess the PXR activation potential of new and existing drugs are becoming integral components of drug discovery programs. PXR is also involved in lipid homeostasis providing opportunities for treatments based on PXR agonists for diseases involving aberrant cholesterol and bile acid levels. The expression of PXR in many other tissues besides liver and intestine suggest PXR may have additional protective functions in the body, which contribute to disease outcomes in diverse clinical situations with potential for novel therapeutic approaches.


Receptors, Steroid/metabolism , Amino Acid Sequence , Animals , Humans , Inactivation, Metabolic , Lipid Metabolism , Mice , Models, Biological , Molecular Sequence Data , Pregnane X Receptor , Receptors, Steroid/chemistry , Receptors, Steroid/genetics , Sequence Homology, Amino Acid , Xenobiotics/metabolism
19.
Clin Cancer Res ; 12(24): 7492-7, 2006 Dec 15.
Article En | MEDLINE | ID: mdl-17189422

PURPOSE: Many chemotherapeutic drugs have an inherent lack of safety due to interindividual variability of hepatic cytochrome P450 (CYP) 3A4 drug metabolism. This reduction in CYP3A4 in cancer patients is possibly mediated by cytokines associated with tumor-derived inflammation. We sought to examine this link by using an explant sarcoma in a novel transgenic mouse model of human CYP3A4 regulation. EXPERIMENTAL DESIGN: Engelbreth-Holm-Swarm sarcoma cells were injected into the hindlimb of transgenic CYP3A4/lacZ mice. Hepatic expression of the human CYP3A4 transgene was analyzed by direct measurement of the reporter gene product, beta-galactosidase enzyme activity. Hepatic expression of murine Cyp3a was analyzed at the mRNA, protein, and function levels. The acute phase response was assessed by examining cytokines [interleukin-6 (IL-6) and tumor necrosis factor] in serum, liver, or tumor as well as hepatic expression of serum amyloid protein P. RESULTS: Engelbreth-Holm-Swarm sarcoma elicited an acute phase response that coincided with down-regulation of the human CYP3A4 transgene in the liver as well as the mouse orthologue Cyp3a11. The reduction of murine hepatic Cyp3a gene expression in tumor-bearing mice resulted in decreased Cyp3a protein expression and consequently a significant reduction in Cyp3a-mediated metabolism of midazolam. Circulating IL-6 was elevated and IL-6 protein was only detected in tumor tissue but not in hepatic tissue. CONCLUSIONS: The current study provides a mechanistic link between cancer-associated inflammation and impaired drug metabolism in vivo. Targeted therapy to reduce inflammation may provide improved clinical benefit for chemotherapy drugs metabolized by hepatic CYP3A4 by improving their pharmacokinetic profile.


Cytochrome P-450 Enzyme System/metabolism , Liver/metabolism , Sarcoma, Experimental/metabolism , Adjuvants, Anesthesia/metabolism , Adjuvants, Anesthesia/pharmacology , Animals , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/genetics , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Liver/enzymology , Liver/pathology , Male , Mice , Mice, Transgenic , Midazolam/metabolism , Midazolam/pharmacology , Sarcoma, Experimental/genetics , Sarcoma, Experimental/pathology , Transgenes , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
20.
Biochem Pharmacol ; 69(4): 709-17, 2005 Feb 15.
Article En | MEDLINE | ID: mdl-15670589

In rodents, high-fat diets promote hepatic lipid accumulation in rodents, activation of peroxisome proliferator activated receptor-alpha (PPARalpha) and upregulation of cytochrome P450 (CYP) 4A gene expression. Lipid-devoid diets containing sucrose and orotic acid (S/OA-diet) also cause lipid infiltration by stimulating intrahepatic lipid synthesis and preventing lipoprotein transport through the Golgi apparatus. This study evaluated the impact of the lipid-deficient S/OA-diet on CYP4A expression and PPARalpha activation in rodent liver. CYP4A protein and laurate omega-hydroxylation activity were increased in rat liver after S/OA-feeding for 21 days. CYP4A1 and CYP4A2 mRNAs were induced to 2.1- and 2.6-fold of control, but mRNAs corresponding to CYP4A3 and the peroxisomal acyl-CoA oxidase (AOX) were unchanged. Coadministration of clofibric acid and the S/OA-diet prevented hepatic lipid accumulation and upregulated CYP4A protein to levels comparable with clofibric acid alone (five-fold of control). Clofibric acid, alone and in combination with the S/OA-diet, upregulated CYP4A1-3 and AOX mRNAs. Hepatic PPARalpha protein was decreased by the S/OA-diet but was increased to 5.7-fold of control by clofibric acid; retinoid X-receptor-alpha (RXRalpha) protein was decreased to 26-41% of control by all treatments. In further studies, administration of the S/OA-diet to control and PPARalpha-null mice promoted hepatic lipid deposition; microsomal CYP4A protein was induced in wild-type but not PPARalpha-null mice. These findings implicate PPARalpha in the induction of CYP4A in rodent liver by the lipid-devoid S/OA-diet. Decreased availability of hepatic PPARalpha and RXRalpha after intake of the diet may contribute to the selective upregulation of hepatic CYP4A1 and CYP4A2 in this model.


Cytochrome P-450 CYP4A/genetics , Microsomes, Liver/enzymology , Orotic Acid/administration & dosage , Sucrose/administration & dosage , Animals , Female , Gene Expression Regulation, Enzymologic , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar , Up-Regulation
...