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










Publication year range
1.
Biol Pharm Bull ; 43(3): 366-374, 2020.
Article in English | MEDLINE | ID: mdl-32115498

ABSTRACT

Selenoprotein P (SeP) is one of the 25 human selenocysteine (Sec)-containing proteins, and is generally thought to function as a plasma carrier of the trace element selenium in the body. Recent studies, however, indicate unsuspected pivotal roles of SeP in human diseases, particularly in type 2 diabetes mellitus (T2DM) and pulmonary arterial hypertension (PAH). In this review, we will summarize the characteristics of SeP and recent advances in the field, especially focusing on the emerging roles of SeP in pathophysiological conditions. We will also discuss potential medical/pharmaceutical applications targeting SeP.


Subject(s)
Selenoprotein P/blood , Selenoprotein P/physiology , Animals , Biomarkers , Diabetes Mellitus, Type 2/physiopathology , Humans , Plasma , Prognosis , Pulmonary Arterial Hypertension/physiopathology , Selenium/metabolism , Selenoprotein P/drug effects
2.
Endocr J ; 66(8): 659-662, 2019 Aug 29.
Article in English | MEDLINE | ID: mdl-31366824

ABSTRACT

Many researchers pay attention to novel secretory factors, such as adipokines or osteokines, secreted by the tissues that were not formerly recognized as classical endocrine organs. The liver also contributes to the onset of various kinds of pathologies of type 2 diabetes and obesity by producing and releasing secretory proteins "hepatokines." By using the information of gene expression in human livers, we rediscovered selenoprotein P (SeP) and leukocyte cell-derived chemotaxin 2 (LECT2) as hepatokines involved in the onset of glucose intolerance. SeP was previously recognized as a selenium transport protein, but we revealed that SeP causes insulin resistance in the muscle and liver. SeP also reduces VEGF signal transduction in vascular endothelial cells, contributing the impaired angiogenesis in diabetes. Importantly, SeP impairs health-promoting effects of exercise training by suppressing reactive oxygen species (ROS)/adenosine monophosphate-dependent protein kinase (AMPK) pathway in the skeletal muscle through its receptor low-density lipoprotein receptor-related protein 1 (LRP1). LECT2, previously-reported as a neutrophil chemotactic protein, promotes skeletal muscle insulin resistance in obesity. Further studies are necessary to develop new diagnostic or therapeutic procedures targeting hepatokines to combat type 2 diabetes or obesity.


Subject(s)
Chemokines/physiology , Diabetes Mellitus, Type 2/etiology , Liver/metabolism , Obesity/etiology , Animals , Chemokines/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Exercise/physiology , Glucose Intolerance/complications , Glucose Intolerance/metabolism , Humans , Insulin Resistance/physiology , Intercellular Signaling Peptides and Proteins/physiology , Muscle, Skeletal/metabolism , Obesity/metabolism , Obesity/therapy , Selenoprotein P/metabolism , Selenoprotein P/physiology
3.
Nat Rev Endocrinol ; 13(9): 509-520, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28621339

ABSTRACT

Hepatic steatosis is an underlying feature of nonalcoholic fatty liver disease (NAFLD), which is the most common form of liver disease and is present in up to ∼70% of individuals who are overweight. NAFLD is also associated with hypertriglyceridaemia and low levels of HDL, glucose intolerance, insulin resistance and type 2 diabetes mellitus. Hepatic steatosis is a strong predictor of the development of insulin resistance and often precedes the onset of other known mediators of insulin resistance. This sequence of events suggests that hepatic steatosis has a causal role in the development of insulin resistance in other tissues, such as skeletal muscle. Hepatokines are proteins that are secreted by hepatocytes, and many hepatokines have been linked to the induction of metabolic dysfunction, including fetuin A, fetuin B, retinol-binding protein 4 (RBP4) and selenoprotein P. In this Review, we describe the factors that influence the development of hepatic steatosis, provide evidence of strong links between hepatic steatosis and insulin resistance in non-hepatic tissues, and discuss recent advances in our understanding of how steatosis alters hepatokine secretion to influence metabolic phenotypes through inter-organ communication.


Subject(s)
Blood Proteins/physiology , Insulin Resistance , Liver/physiopathology , Non-alcoholic Fatty Liver Disease/physiopathology , Animals , Diabetes Mellitus, Type 2 , Diet, High-Fat , Fetuin-B/physiology , Humans , Hypertriglyceridemia , Lipid Metabolism/physiology , Obesity , Overweight , Retinol-Binding Proteins, Plasma/physiology , Selenoprotein P/physiology , alpha-2-HS-Glycoprotein/physiology
4.
Acta Biochim Pol ; 63(3): 459-67, 2016.
Article in English | MEDLINE | ID: mdl-27262842

ABSTRACT

Nowadays non-alcoholic fatty liver disease (NAFLD) is becoming the most common chronic liver pathology both in adults and children. NAFLD manifestation ranges from a simple liver steatosis to steatohepatitis (nonalcoholic steatohepatitis - NASH), which may progress to advanced fibrosis, cirrhosis and end-stage liver disease. Due to the coexistence of visceral obesity, insulin resistance and dyslipidemia, NAFLD is considered to be the hepatic manifestation of metabolic syndrome. In recent years, in the pathogenesis of metabolic syndrome, type 2 diabetes mellitus, cardiovascular disease and also NAFLD, more and more attention has been paid to the so-called organokines, proteins with both paracrine or/and endocrine activities. These include most known adipokines (mainly produced by adipose tissue), myokines (mainly produced by skeletal muscles) and hepatokines exclusively or predominantly produced by the liver. It was shown that the liver may affect the lipids and glucose metabolism by hepatokines released into the blood and NAFLD seems to be associated with altered hepatokines production. Fetuin-A, fibroblast growth factor-21 (FGF-21), selenoprotein P, sex hormone-binding globulin (SHBG), angiopoietin-related growth factor (also known as angiopoietin-related protein 6) and leukocyte derived chemotaxin 2 (LECT2) are considered as the most important hepatokines. In this review, we provide an overview of the main hepatokines and we summarize the association of liver-derived proteins with the development and progression of NAFLD.


Subject(s)
Non-alcoholic Fatty Liver Disease/metabolism , Animals , Fibroblast Growth Factors/physiology , Humans , Intercellular Signaling Peptides and Proteins/physiology , Liver/metabolism , Liver/pathology , Selenoprotein P/physiology , Sex Hormone-Binding Globulin/physiology , Signal Transduction , alpha-2-HS-Glycoprotein/physiology
5.
J Clin Invest ; 125(7): 2646-60, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26053663

ABSTRACT

Patients with inflammatory bowel disease are at increased risk for colon cancer due to augmented oxidative stress. These patients also have compromised antioxidant defenses as the result of nutritional deficiencies. The micronutrient selenium is essential for selenoprotein production and is transported from the liver to target tissues via selenoprotein P (SEPP1). Target tissues also produce SEPP1, which is thought to possess an endogenous antioxidant function. Here, we have shown that mice with Sepp1 haploinsufficiency or mutations that disrupt either the selenium transport or the enzymatic domain of SEPP1 exhibit increased colitis-associated carcinogenesis as the result of increased genomic instability and promotion of a protumorigenic microenvironment. Reduced SEPP1 function markedly increased M2-polarized macrophages, indicating a role for SEPP1 in macrophage polarization and immune function. Furthermore, compared with partial loss, complete loss of SEPP1 substantially reduced tumor burden, in part due to increased apoptosis. Using intestinal organoid cultures, we found that, compared with those from WT animals, Sepp1-null cultures display increased stem cell characteristics that are coupled with increased ROS production, DNA damage, proliferation, decreased cell survival, and modulation of WNT signaling in response to H2O2-mediated oxidative stress. Together, these data demonstrate that SEPP1 influences inflammatory tumorigenesis by affecting genomic stability, the inflammatory microenvironment, and epithelial stem cell functions.


Subject(s)
Colitis/complications , Colonic Neoplasms/etiology , Selenoprotein P/physiology , Animals , Antioxidants/metabolism , Apoptosis , Colonic Neoplasms/pathology , Colonic Neoplasms/physiopathology , DNA Damage , Genomic Instability , Haploinsufficiency , Macrophages/classification , Macrophages/pathology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Site-Directed , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/physiology , Oxidative Stress , Protein Structure, Tertiary , Selenium/administration & dosage , Selenium/metabolism , Selenoprotein P/deficiency , Selenoprotein P/genetics , Tumor Microenvironment , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology
6.
FASEB J ; 28(8): 3579-88, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24760755

ABSTRACT

Selenoprotein P (Sepp1) and its receptor, apolipoprotein E receptor 2 (apoER2), account for brain retaining selenium better than other tissues. The primary sources of Sepp1 in plasma and brain are hepatocytes and astrocytes, respectively. ApoER2 is expressed in varying amounts by tissues; within the brain it is expressed primarily by neurons. Knockout of Sepp1 or apoER2 lowers brain selenium from ∼120 to ∼50 ng/g and leads to severe neurodegeneration and death in mild selenium deficiency. Interactions of Sepp1 and apoER2 that protect against this injury have not been characterized. We studied Sepp1, apoER2, and brain selenium in knockout mice. Immunocytochemistry showed that apoER2 mediates Sepp1 uptake at the blood-brain barrier. When Sepp1(-/-) or apoER2(-/-) mice developed severe neurodegeneration caused by mild selenium deficiency, brain selenium was ∼35 ng/g. In extreme selenium deficiency, however, brain selenium of ∼12 ng/g was tolerated when both Sepp1 and apoER2 were intact in the brain. These findings indicate that tandem Sepp1-apoER2 interactions supply selenium for maintenance of brain neurons. One interaction is at the blood-brain barrier, and the other is within the brain. We postulate that Sepp1 inside the blood-brain barrier is taken up by neurons via apoER2, concentrating brain selenium in them.


Subject(s)
Blood-Brain Barrier/physiology , Brain/metabolism , LDL-Receptor Related Proteins/physiology , Nerve Degeneration/prevention & control , Selenium/metabolism , Selenoprotein P/physiology , Animals , Animals, Congenic , Biological Transport , Brain/embryology , Brain/growth & development , Capillaries/metabolism , Choroid Plexus/embryology , Choroid Plexus/growth & development , Choroid Plexus/metabolism , Endocytosis , Endothelial Cells/metabolism , Female , LDL-Receptor Related Proteins/deficiency , Low Density Lipoprotein Receptor-Related Protein-2/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/etiology , Nerve Degeneration/metabolism , Neurons/metabolism , Pregnancy , Selenium/administration & dosage , Selenium/deficiency , Selenium/pharmacokinetics , Selenoprotein P/deficiency
7.
Metallomics ; 6(5): 1043-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24626785

ABSTRACT

Selenium (Se) plays an important role in bone physiology as best reflected by Kashin-Beck disease, an endemic Se-dependent osteoarthritis. Bone development is delayed in children with mutations in SECIS binding protein 2 (SBP2), a central factor for selenoprotein biosynthesis. Circulating selenoprotein P (SePP) is positively associated with bone turnover in humans, yet its function for bone homeostasis is not known. We have analysed murine models of altered Se metabolism. Most of the known selenoprotein genes and factors needed for selenoprotein biosynthesis are expressed in bones. Bone Se is not associated with the mineral but exclusively with the organic matrix. Genetic ablation of Sepp-expression causes a drastic decline in serum (25-fold) but only a mild reduction in bone (2.5-fold) Se concentrations. Cell-specific expression of a SePP transgene in hepatocytes efficiently restores bone Se levels in Sepp-knockout mice. Of the two known SePP receptors, Lrp8 was detected in bones while Lrp2 was absent. Interestingly, Lrp8 mRNA concentrations were strongly increased in bones of Sepp-knockout mice likely in order to counteract the developing Se deficiency. Our data highlight SePP as the essential Se transporter to bones, and suggest a novel feedback mechanism for preferential uptake of Se in Se-deprived bones, thereby contributing to our understanding of hepatic osteodystrophy and the consistent bone phenotype observed in subjects with inherited selenoprotein biosynthesis mutations.


Subject(s)
Bone and Bones/metabolism , Carrier Proteins/metabolism , Selenium/metabolism , Selenoprotein P/physiology , Animals , Blotting, Western , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Selenoprotein P/genetics
8.
Int J Radiat Oncol Biol Phys ; 87(3): 619-25, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24074935

ABSTRACT

PURPOSE: Radiation is a common mode of cancer therapy whose outcome is often limited because of normal tissue toxicity. We have shown previously that the accumulation of radiation-induced late reactive oxygen species (ROS) precedes cell death, suggesting that metabolic oxidative stress could regulate cellular radiation response. The purpose of this study was to investigate whether selenoprotein P (SEPP1), a major supplier of selenium to tissues and an antioxidant, regulates late ROS accumulation and toxicity in irradiated normal human fibroblasts (NHFs). METHODS AND MATERIALS: Flow cytometry analysis of cell viability, cell cycle phase distribution, and dihydroethidium oxidation, along with clonogenic assays, were used to measure oxidative stress and toxicity. Human antioxidant mechanisms array and quantitative real-time polymerase chain reaction assays were used to measure gene expression during late ROS accumulation in irradiated NHFs. Sodium selenite addition and SEPP1 overexpression were used to determine the causality of SEPP1 regulating late ROS accumulation and toxicity in irradiated NHFs. RESULTS: Irradiated NHFs showed late ROS accumulation (4.5-fold increase from control; P<.05) that occurs after activation of the cell cycle checkpoint pathways and precedes cell death. The mRNA levels of CuZn- and Mn-superoxide dismutase, catalase, peroxiredoxin 3, and thioredoxin reductase 1 increased approximately 2- to 3-fold, whereas mRNA levels of cold shock domain containing E1 and SEPP1 increased more than 6-fold (P<.05). The addition of sodium selenite before the radiation treatment suppressed toxicity (45%; P<.05). SEPP1 overexpression suppressed radiation-induced late ROS accumulation (35%; P<.05) and protected NHFs from radiation-induced toxicity (58%; P<.05). CONCLUSION: SEPP1 mitigates radiation-induced late ROS accumulation and normal cell injury.


Subject(s)
Radiation Injuries/prevention & control , Reactive Oxygen Species/metabolism , Selenoprotein P/physiology , Cell Cycle Checkpoints/physiology , Cell Cycle Checkpoints/radiation effects , Cell Death , Cell Survival , Dose-Response Relationship, Radiation , Ethidium/analogs & derivatives , Ethidium/metabolism , Fibroblasts/metabolism , Fibroblasts/radiation effects , Genes, vif , Humans , Oxidative Stress/genetics , Real-Time Polymerase Chain Reaction , Selenoprotein P/genetics , Selenoprotein P/metabolism , Sodium Selenite/pharmacology
9.
Metallomics ; 5(7): 861-70, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23652332

ABSTRACT

Aggregation and cytotoxicity of the amyloid-ß (Aß) peptide with transition metal ions in neuronal cells have been suggested to be involved in the progression of Alzheimer's disease (AD). A therapeutic strategy to combat this incurable disease is to design chemical agents to target metal-Aß species. Selenoproteins are a group of special proteins that contain the 21st amino acid Sec in their sequence. Due to the presence of Sec, studies of this group of proteins are basically focused on their roles in regulating redox potential and scavenging reactive oxygen species. Here, we reported that the His-rich domain of selenoprotein P (SelP-H) and the Sec-to-Cys mutant selenoprotein M (SelM') are capable of binding transition metal ions and modulating the Zn(2+)-mediated Aß aggregation, ROS production and neurotoxicity. SelM' (U48C) and SelP-H were found to coordinate 0.5 and 2 molar equivalents of Zn(2+)/Cd(2+) with micromolar and submicromolar affinities, respectively. Metal binding induced the structural changes in SelP-H and SelM' according to the circular dichorism spectra. Zn(2+) binding to Aß42 almost completely suppressed Aß42 fibrillization, which could be significantly restored by SelP-H and SelM', as observed by thioflavin T (ThT) fluorescence and transmission electron microscopy (TEM). Interestingly, both SelP-H and SelM' inhibited Zn(2+)-Aß42-induced neurotoxicity and the intracellular ROS production in living cells. These studies suggest that SelP and SelM may play certain roles in regulating redox balance as well as metal homeostasis.


Subject(s)
Amyloid beta-Peptides/metabolism , Selenoprotein P/physiology , Selenoproteins/physiology , Zinc/metabolism , Amino Acid Sequence , Amyloid beta-Peptides/toxicity , Base Sequence , Circular Dichroism , DNA Primers , Escherichia coli/metabolism , Microscopy, Electron, Transmission , Molecular Sequence Data , Selenoprotein P/chemistry , Selenoprotein P/metabolism , Selenoproteins/chemistry , Selenoproteins/metabolism , Sequence Homology, Amino Acid , Spectrophotometry, Ultraviolet
11.
Am J Physiol Endocrinol Metab ; 300(1): E77-85, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20959537

ABSTRACT

Oxidative stress and low-grade inflammation have been implicated in obesity and insulin resistance. As a selenium transporter, ubiquitously expressed selenoprotein P (SeP) is known to play a role in the regulation of antioxidant enzyme activity. However, SeP expression and regulation in adipose tissue in obesity and its role in inflammation and adipocyte biology remain unexplored. In this study, we examined Sepp1 gene expression and regulation in adipose tissue of obese rodents and characterized the role of Sepp1 in adipose inflammation and adipogenesis in 3T3-L1 adipocytes. We found that Sepp1 gene expression was significantly reduced in adipose tissue of ob/ob and high-fat diet-induced obese mice as well as in primary adipose cells isolated from Zucker obese rats. Rosiglitazone administration increased SeP protein expression in adipose tissue of obese mice. Treatment of either TNFα or H(2)O(2) significantly reduced Sepp1 gene expression in a time- and dose-dependent manner in 3T3-L1 adipocytes. Interestingly, Sepp1 gene silencing resulted in the reduction in glutathione peroxidase activity and the upregulation of inflammatory cytokines MCP-1 and IL-6 in preadipocytes, leading to the inhibition of adipogenesis and adipokine and lipogenic gene expression. Most strikingly, coculturing Sepp1 KD cells resulted in a marked inhibition of normal 3T3-L1 adipocyte differentiation. We conclude that SeP has an important role in adipocyte differentiation via modulating oxidative stress and inflammatory response.


Subject(s)
Adipogenesis , Adipose Tissue, White/metabolism , Down-Regulation , Inflammation Mediators/metabolism , Obesity/metabolism , Oxidative Stress , Selenoprotein P/physiology , 3T3-L1 Cells , Adipose Tissue, White/cytology , Adipose Tissue, White/drug effects , Animals , Cells, Cultured , Coculture Techniques , Down-Regulation/drug effects , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Obese , RNA, Messenger/metabolism , RNA, Small Interfering , Rats , Rats, Zucker , Selenoprotein P/genetics
12.
Ethn Dis ; 20(1 Suppl 1): S1-92-5, 2010.
Article in English | MEDLINE | ID: mdl-20521393

ABSTRACT

INTRODUCTION: Selenoprotein P (SelP) plays a critical role in neuronal survival and is associated with Alzheimer's pathology. We sought to determine a potential neuroprotective role for SelP in Alzheimer's disease. METHODS: We utilized RNAi to reduce SelP expression in neuronal N2A cells, and determined cell viability with flow cytometry. We subsequently measured neurotoxicity from exposure of aggregated amyloid beta (Abeta) peptides to SelP-knockdown and control N2A cells. RESULTS: We found that knockdown of SelP using siRNA in N2A cells reduced viability and increased apoptotic cell death. Additionally, knockdown of SelP using siRNA in N2A cells resulted in increased AB toxicity. CONCLUSIONS: Our findings demonstrate that SelP protects neuronal cells from Abeta-induced toxicity, suggesting a neuroprotective role for SelP in preventing neurodegenerative disorders.


Subject(s)
Alzheimer Disease/physiopathology , Oxidative Stress/physiology , Selenoprotein P/physiology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cell Death/physiology , Gene Expression , Gene Knockdown Techniques , Humans , In Situ Nick-End Labeling , RNA, Small Interfering/genetics , Selenoprotein P/metabolism , Transfection
13.
PLoS One ; 5(3): e9911, 2010 Mar 29.
Article in English | MEDLINE | ID: mdl-20360971

ABSTRACT

The ocular surface is always attacked by oxidative stress, and cornea epithelial cells are supposed to have their own recovery system against oxidative stress. Therefore we hypothesized that tears supply key molecules for preventing oxidative stress in cornea. The potential target key molecule we focused is selenoprotein P (SeP). SeP is a carrier of selenium, which is an essential trace element for many animals, for oxidative stress metabolism in the organism, and was extremely expressed in lacrimal gland. An experiment was performed with SeP eye drops in a rat dry eye model, prepared by removing the lacrimal glands. The anticipated improvement in corneal dry eye index and the suppression of oxidative stress markers were observed in SeP eye drop group. Furthermore, the concentration of SeP was significantly higher in dry eye patients compared with normal volunteers. Collectively, we concluded that tear SeP is a key molecule to protect the ocular surface cells against environmental oxidative stress.


Subject(s)
Cornea/metabolism , Gene Expression Regulation , Lacrimal Apparatus/metabolism , Oxidative Stress , Selenoprotein P/physiology , Tears/metabolism , Animals , Disease Models, Animal , Dry Eye Syndromes/metabolism , Humans , Male , Models, Biological , Rats , Rats, Sprague-Dawley , Selenium/pharmacology
14.
Biochim Biophys Acta ; 1790(11): 1441-7, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19345254

ABSTRACT

Selenoprotein P (Sepp1) is a secreted protein that is made up of 2 domains. The larger N-terminal domain contains 1 selenocysteine residue in a redox motif and the smaller C-terminal domain contains the other 9 selenocysteines. Sepp1 isoforms of varying lengths occur but quantitation of them has not been achieved. Hepatic synthesis of Sepp1 affects whole-body selenium content and the liver is the source of most plasma Sepp1. ApoER2, a member of the lipoprotein receptor family, binds Sepp1 and facilitates its uptake into the testis and retention of its selenium by the brain. Megalin, another lipoprotein receptor, facilitates uptake of filtered Sepp1 into proximal tubule cells of the kidney. Thus, Sepp1 serves in homeostasis and distribution of selenium. Mice with deletion of Sepp1 suffer greater morbidity and mortality from infection with Trypanosoma congolense than do wild-type mice. Mice that express only the N-terminal domain of Sepp1 have the same severity of illness as wild-type mice, indicating that the protective function of Sepp1 against the infection resides in the N-terminal (redox) domain. Thus, Sepp1 has several functions. In addition, plasma Sepp1 concentration falls in selenium deficiency and, therefore, it can be used as an index of selenium nutritional status.


Subject(s)
Mammals/genetics , Selenoprotein P/genetics , Selenoprotein P/physiology , Animals , Biological Transport/genetics , Biological Transport/physiology , Brain Injuries/genetics , Brain Injuries/metabolism , Homeostasis/genetics , Humans , Male , Mammals/metabolism , Mammals/physiology , Mice , Models, Biological , Parasitic Diseases/genetics , Parasitic Diseases/metabolism , Selenium/metabolism , Selenoprotein P/metabolism , Spermatogenesis/genetics
15.
Biochim Biophys Acta ; 1790(11): 1546-54, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19289153

ABSTRACT

Selenoproteins comprise a unique class of proteins that contain selenium in the form of selenocysteine. Several selenoproteins have been implicated in the risk or development of cancers in humans by genetic data. These include Selenoprotein P, 3 members of the glutathione peroxidase family of anti-oxidant enzymes and Sep15. At-risk alleles in the germline indicate a likely role in determining susceptibility to cancer, while loss of heterozygosity or chromosomal epigenetic silencing indicate that the reduction in the levels of the corresponding proteins contribute to malignant progression. Lower levels of these proteins are likely to be detrimental due to the resulting cellular stress and perturbations in important regulatory signaling pathways. The genetic data indicating the involvement of these selenoproteins in cancer etiology are discussed, as are the possible mechanisms by which these genes might promote carcinogenesis.


Subject(s)
Neoplasms/genetics , Selenoproteins/physiology , Animals , Disease Models, Animal , Disease Progression , Genetic Predisposition to Disease , Glutathione Peroxidase/genetics , Glutathione Peroxidase/physiology , Humans , Mice , Risk , Selenoprotein P/genetics , Selenoprotein P/physiology , Selenoproteins/genetics
16.
Nucleic Acids Res ; 35(12): 3963-73, 2007.
Article in English | MEDLINE | ID: mdl-17553827

ABSTRACT

Selenoprotein P (Sel P) is a selenium-rich glycoprotein believed to play a key role in selenium (Se) transport throughout the body. Development of a Sel P knockout mouse model has supported this notion and initial studies have indicated that selenium supply to various tissues is differentially affected by genetic deletion of Sel P. Se in the form of the amino acid, selenocysteine, is incorporated into selenoproteins at UGA codons. Thus, Se availability affects not only selenoprotein levels, but also the turnover of selenoprotein mRNAs via the nonsense-mediated decay pathway. We investigated how genetic deletion of Sel P in mice affected levels of the mRNAs encoding all known members of the murine selenoprotein family, as well as three non-selenoprotein factors involved in their synthesis, selenophosphate synthetase 1 (SPS1), SECIS-binding protein 2 (SBP2) and SECp43. Our findings present a comprehensive description of selenoprotein mRNA expression in the following murine tissues: brain, heart, intestine, kidney, liver, lung, spleen and testes. We also describe how abundance of selenoproteins and selenoprotein-synthesis factors are affected by genetic deletion of Sel P in some of these tissues, providing insight into how the presence of this selenoprotein influences selenoprotein mRNA levels, and thus, the selenoproteome.


Subject(s)
Selenium/metabolism , Selenoprotein P/genetics , Selenoproteins/metabolism , Animals , Brain/metabolism , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Proteome/metabolism , RNA, Messenger/metabolism , Selenoprotein P/physiology , Selenoproteins/genetics , Testis/metabolism , Tissue Distribution
17.
J Biol Chem ; 282(16): 12290-7, 2007 Apr 20.
Article in English | MEDLINE | ID: mdl-17314095

ABSTRACT

Selenium is a micronutrient that is essential for the production of normal spermatozoa. The selenium-rich plasma protein selenoprotein P (Sepp1) is required for maintenance of testis selenium and for fertility of the male mouse. Sepp1 trafficking in the seminiferous epithelium was studied using conventional methods and mice with gene deletions. Immunocytochemistry demonstrated that Sepp1 is present in vesicle-like structures in the basal region of Sertoli cells, suggesting that the protein is taken up intact. Sepp1 affinity chromatography of a testicular extract followed by mass spectrometry-based identification of bound proteins identified apolipoprotein E receptor 2 (ApoER2) as a candidate testis Sepp1 receptor. In situ hybridization analysis identified Sertoli cells as the only cell type in the seminiferous epithelium with detectable ApoER2 expression. Testis selenium levels in apoER2(-/-) males were sharply reduced from those in apoER2(+/+) males and were comparable with the depressed levels found in Sepp1(-/-) males. However, liver selenium levels were unchanged by deletion of apoER2. Immunocytochemistry did not detect Sepp1 in the Sertoli cells of apoER2(-/-) males, consistent with a defect in the receptor-mediated Sepp1 uptake pathway. Phase contrast microscopy revealed identical sperm defects in apoER2(-/-) and Sepp1(-/-) mice. Co-immunoprecipitation analysis demonstrated an interaction of testis ApoER2 with Sepp1. These data demonstrate that Sertoli cell ApoER2 is a Sepp1 receptor and a component of the selenium delivery pathway to spermatogenic cells.


Subject(s)
Receptors, Lipoprotein/physiology , Selenium/pharmacology , Selenoprotein P/physiology , Spermatozoa/metabolism , Testis/metabolism , Amino Acid Sequence , Animals , LDL-Receptor Related Proteins , Male , Mice , Mice, Transgenic , Microscopy, Phase-Contrast , Molecular Sequence Data , Phenotype , Rats , Seminiferous Epithelium/metabolism
18.
Biochim Biophys Acta ; 1760(12): 1789-93, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17014962

ABSTRACT

Deletion of the mouse selenoprotein P gene (Sepp1) lowers selenium concentrations in many tissues. We examined selenium homeostasis in Sepp1(-/-) and Sepp1(+/+) mice to assess the mechanism of this. The liver produces and exports selenoprotein P, which transports selenium to peripheral tissues, and urinary selenium metabolites, which regulate whole-body selenium. At intakes of selenium near the nutritional requirement, Sepp1(-/-) mice had whole-body selenium concentrations 72 to 75% of Sepp1(+/+) mice. Genotype did not affect dietary intake of selenium. Sepp1(-/-) mice excreted in their urine approximately 1.5 times more selenium in relation to their whole-body selenium than did Sepp1(+/+) mice. In addition, Sepp1(-/-) mice gavaged with (75)SeO(2-)(3) excreted 1.7 to 2.4 times as much of the (75)Se in the urine as did Sepp1(+/+) mice. These findings demonstrate that deletion of selenoprotein P raises urinary excretion of selenium. When urinary small-molecule (75)Se was injected intravenously into mice, over 90% of the (75)Se appeared in the urine within 24 h, regardless of selenium status. This shows that urinary selenium is dedicated to excretion and not to utilization by tissues. Our results indicate that deletion of selenoprotein P leads to increased urinary selenium excretion. We propose that the absence of selenoprotein P synthesis in the liver makes more selenium available for urinary metabolite synthesis, increasing loss of selenium from the organism and causing the decrease in whole-body selenium and some of the decreases observed in tissues of Sepp1(-/-) mice.


Subject(s)
Selenium/urine , Selenoprotein P/physiology , Up-Regulation/physiology , Animals , Male , Mice , Mice, Knockout , Selenium/pharmacokinetics , Selenoprotein P/genetics
19.
Curr Opin Clin Nutr Metab Care ; 9(3): 233-8, 2006 May.
Article in English | MEDLINE | ID: mdl-16607122

ABSTRACT

PURPOSE OF REVIEW: To review the reason for and clinical effects of selenium supplementation in critically ill patients. RECENT FINDINGS: Selenium-dependent enzymes and selenoprotein P regulate immune and endothelial cell function. Obviously not the anorganic compounds of selenium but the activity of selenium-dependent enzymes is the most important factor modulating the immune system and the clinical outcome of patients. Despite low selenium levels in severely ill patients and low glutathione peroxidase activity associated with the extent of multiorgan dysfunction, only a few trials have investigated the effect of selenium supplementation on clinical outcome. A metaanalysis did not reveal a statistically significant survival rate with selenium supplementation, but suggested a dose-dependent trend. The recently completed multicentre trial on high-dose selenium supplementation in septic patients also did not reveal a significant overall reduction in mortality. SUMMARY: The available evidence suggests that selenoproteins play an important role in the immunomodulation of critically ill patients and a sodium selenite supplementation upregulates these selenoenzymes. The intervention trials with sodium selenite performed to date are small and therefore only a tendency in reduction of morbidity and mortality could be demonstrated. Larger trials are necessary to show the supposed benefits and risks of selenite supplementation in critically ill patients.


Subject(s)
Antioxidants/therapeutic use , Critical Illness/therapy , Selenium/metabolism , Selenium/therapeutic use , Selenoproteins/immunology , Antioxidants/metabolism , Critical Illness/mortality , Dietary Supplements , Dose-Response Relationship, Drug , Dose-Response Relationship, Immunologic , Humans , Selenoprotein P/immunology , Selenoprotein P/metabolism , Selenoprotein P/physiology , Selenoproteins/metabolism , Selenoproteins/physiology , Treatment Outcome
20.
Wei Sheng Yan Jiu ; 33(5): 572-4, 2004 Sep.
Article in Chinese | MEDLINE | ID: mdl-15612483

ABSTRACT

OBJECTIVE: To probe the function and structure of human selenoprotein P (SELP). METHODS: Several bioinformatic server are used to analyze exons, hydrophobic nature, transmembrane helices and the bonding state of cysteines respectively, as well as the secondary structure and relative solvent accessibility. The homology analysis and searching for motifs or domains in SELP are also carried out. RESULTS: Two transmembrane helices are found, as is consistent with the hydrophobic nature of SELP. Matured SELP consists of four modules: M1 (Glu20-Ser67/Lys68), M2 (Ser67/Lys68-Asp138/Arg139), M3(Asp138/Arg139-Thr178), M4 (Thr179-Asn381). A coiled coil exists in the second module whose regular secondary structure is higher than that of other three. The percentage of loop structure is higher in the first, third and fourth modules, so they may be three reactive center. The fourth module is the most active one which has a histidine-rich region (His204-His257) on the surface of SELP. Because most of the residues are the E type of solvent accessibility, SELP appears not to be globular. SELP consists of two kinds of domain, so it more probably has more than one function. HUNB_SCAAL, ZP12_BRARE, BR11_BRARE, BRN1_HUMAN which play roles in the development of brain are found have sequence similarity with SELP and the similarity may be the specificity to brain. Other two similar proteins are KNOB_PLAFG and HPN_ HELPY which anchor host thrombospondin or a parasite analog in a binding complex with the endothelial cell receptor and strongly binds nickel and zinc respectively. CONCLUSION: The results showed suggest that SELP may have metal binding and endothelial cell membrane anchoring activity which are backed up not only by homology analysis but also the prediction of structure. Moreover, the results also suggest the specificity of SELP to brain.


Subject(s)
Selenoprotein P/chemistry , Selenoprotein P/physiology , Amino Acid Sequence , Brain/growth & development , Databases, Protein , Endothelial Cells/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Nickel/metabolism , Protein Structure, Secondary , Sequence Homology, Amino Acid , Zinc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...