Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Breast Cancer Res Treat ; 201(2): 317-328, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37378696

ABSTRACT

PURPOSE: HER2-positive breast cancer has a high chance of achieving pathological complete response when HSD17B4, responsible for peroxisomal ß-oxidation of very long-chain fatty acids (VLCFA) and estradiol, is methylation-silenced. Here, we aimed to identify the underlying molecular mechanism. METHODS: Using a HER2-positive breast cancer cell line, BT-474, control and knock-out (KO) clones were obtained. Metabolic characteristics were analyzed using a Seahorse Flux analyzer. RESULTS: HSD17B4 KO suppressed cellular proliferation, and enhanced sensitivity to lapatinib approximately tenfold. The KO led to accumulation of VLCFA and a decrease of polyunsaturated fatty acids (PUFAs), such as docosahexaenoic acid (DHA) and arachidonic acid. HSD17B4 KO increased Akt phosphorylation, possibly via decreased DHA, and genes involved in oxidative phosphorylation (OxPhos) and electron transport chain (ETC) were upregulated. Increased mitochondrial ATP production in the KO cells was confirmed by extracellular flux analyzer. Increased OxPhos led to severe dependence of the KO cells on pyruvate from glycolysis. Suppression of glycolysis by lapatinib led to severe delayed suppression of OxPhos in KO cells. CONCLUSION: HSD17B4 KO in BT-474 cells caused a decrease of PUFAs, increased Akt phosphorylation, enhanced glucose dependence of OxPhos, and increased sensitivity to inhibition of HER2, upstream of Akt. This mechanism may be applicable to other HER2-positive glucose-dependent breast cancer cells with HSD17B4 silencing.


Subject(s)
Breast Neoplasms , Humans , Female , Lapatinib/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Methylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Glucose , Cell Line, Tumor , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Peroxisomal Multifunctional Protein-2/genetics , Peroxisomal Multifunctional Protein-2/metabolism
2.
Mol Cells ; 44(4): 214-222, 2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33935042

ABSTRACT

Phosphatidylserine (PS), a negatively charged phospholipid exclusively located in the inner leaflet of the plasma membrane, is involved in various cellular processes such as blood coagulation, myoblast fusion, mammalian fertilization, and clearance of apoptotic cells. Proteins that specifically interact with PS must be identified to comprehensively understand the cellular processes involving PS. However, only a limited number of proteins are known to associate with PS. To identify PS-associating proteins, we performed a pulldown assay using streptavidin-coated magnetic beads on which biotin-linked PS was immobilized. Using this approach, we identified Hsd17b4, a peroxisomal protein, as a PS-associating protein. Hsd17b4 strongly associated with PS, but not with phosphatidylcholine or sphingomyelin, and the Scp-2-like domain of Hsd17b4 was responsible for this association. The association was disrupted by PS in liposomes, but not by free PS or the components of PS. In addition, translocation of PS to the outer leaflet of the plasma membrane enriched Hsd17b4 in peroxisomes. Collectively, this study suggests an unexpected role of PS as a regulator of the subcellular localization of Hsd17b4.


Subject(s)
Peroxisomal Multifunctional Protein-2/metabolism , Peroxisomes/metabolism , Phosphatidylserines/metabolism , Animals , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL
3.
Int J Mol Sci ; 22(6)2021 Mar 13.
Article in English | MEDLINE | ID: mdl-33805726

ABSTRACT

An electromagnetic field (EMF) may affect the functions of uterine tissues. This study hypothesized that EMF changes the estrogenic activity of pig myometrium during the peri-implantation period. Tissue was collected on days 15-16 of the gestation and incubated in the presence of EMF (50 and 120 Hz, 2 and 4 h). The cytochrome P450 aromatase type 3 (CYP19A3) and hydroxysteroid 17ß dehydrogenase type 4 (HSD17B4) mRNA transcript abundance, cytochrome P450arom (aromatase), and 17ß hydroxysteroid dehydrogenase 17ßHSD) protein abundance and estrone (E1) and estradiol-17ß (E2) release were examined using Real-Time PCR, Western blot and radioimmunoassay. Selected myometrial slices were treated with progesterone (P4) to determine whether it functions as a protector against EMF. CYP19A3 mRNA transcript abundance in slices treated with EMF was less at 50 Hz (2 h) and greater at 120 Hz (2 and 4 h). HSD17B4 mRNA transcript was greater in slices treated with EMF at 120 Hz (2 h). Progesterone diminished EMF-related effects on CYP19A3 and HSD17B4. When P4 was added, EMF had suppressive (50 and 120 Hz, 2 h) or enhancing (50 Hz, 4 h) effects on aromatase abundance. The E1 release was lower after 4 h of EMF treatment at 50 Hz and P4 did not protect myometrial E1 release. In conclusion, EMF alters the synthesis and release of E1 and did not affect E2 release in the myometrium during the peri-implantation period.


Subject(s)
Electromagnetic Fields/adverse effects , Embryo Implantation/radiation effects , Estradiol/metabolism , Estrone/metabolism , Gene Expression Regulation/radiation effects , Myometrium/radiation effects , Animals , Aromatase/genetics , Aromatase/metabolism , Electromagnetic Radiation , Female , Myometrium/metabolism , Peroxisomal Multifunctional Protein-2/genetics , Peroxisomal Multifunctional Protein-2/metabolism , Progesterone/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Swine , Tissue Culture Techniques
4.
Cancer Res ; 81(13): 3593-3606, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33762355

ABSTRACT

Molecular mechanisms underlying intratumoral androgenesis and aberrant androgen receptor (AR) activation in prostate cancer remain poorly understood. Here we demonstrate that ectopic expression of the E3 ubiquitin ligase adaptor speckle-type poxvirus and zinc finger domain protein (SPOP) stabilizes 17ßHSD4. SPOP bound a functional substrate-binding consensus (SBC) motif 315RATST319 in 17ßHSD4 and promoted nondegradable K27- and K29-linked polyubiquitination of 17ßHSD4. The effect of SPOP was antagonized by serum- and glucocorticoid kinase-3 (SGK3)-mediated phosphorylation of serine 318 (S318) in the SBC and S318 phosphorylation-dependent binding of SKP2 E3 ligase and subsequent K48-linked polyubiquitination and proteasomal degradation of 17ßHSD4. Prostate cancer-associated SPOP mutations impaired the SPOP-17ßHSD4 interaction, caused 17ßHSD4 protein destruction in prostate cancer cells in culture and patient specimens, and increased testosterone production and prostate cancer cell growth in vitro and in mouse models. Thus, we have identified SPOP and SKP2 as two essential E3 ubiquitin ligases that exert opposite effects on 17ßHSD4 protein degradation and intratumoral androgenesis in prostate cancer cells. We further demonstrate that SPOP mutations or SKP2 overexpression contribute to prostate cancer progression by decreasing 17ßHSD4 expression and increasing intratumoral androgen synthesis. SIGNIFICANCE: This study reveals a novel mechanism of aberrant AR activation in SPOP-mutated prostate cancer and uncovers putative biomarkers for effective treatment by AR-targeted therapies.


Subject(s)
Androgens/metabolism , Gene Expression Regulation, Neoplastic , Mutation , Nuclear Proteins/metabolism , Peroxisomal Multifunctional Protein-2/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Repressor Proteins/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Humans , Male , Mice , Mice, SCID , Nuclear Proteins/genetics , Peroxisomal Multifunctional Protein-2/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Proteolysis , Receptors, Androgen/genetics , Repressor Proteins/genetics , Tumor Cells, Cultured , Ubiquitination , Xenograft Model Antitumor Assays
5.
PLoS One ; 16(2): e0246279, 2021.
Article in English | MEDLINE | ID: mdl-33529214

ABSTRACT

Tail adipose as one of the important functional tissues can enhance hazardous environments tolerance for sheep. The objective of this study was to gain insight into the underlying development mechanisms of this trait. A quantitative analysis of protein abundance in ovine tail/rump adipose tissue was performed between Chinese local fat- (Kazakh, Hu and Lanzhou) and thin-tailed (Alpine Merino, Tibetan) sheep in the present study by using lable-free approach. Results showed that 3400 proteins were identified in the five breeds, and 804 were differentially expressed proteins, including 638 up regulated proteins and 83 down regulated proteins in the tail adipose tissues between fat- and thin-tailed sheep, and 8 clusters were distinguished for all the DEPs' expression patterns. The differentially expressed proteins are mainly associated with metabolism pathways and peroxisome proliferator activated receptor signaling pathway. Furthermore, the proteomics results were validated by quantitative real-time PCR and Western Blot. Our research has also suggested that the up-regulated proteins ACSL1, HSD17ß4, FABP4 in the tail adipose tissue might contribute to tail fat deposition by facilitating the proliferation of adipocytes and fat accumulation in tail/rump of sheep. Particularly, FABP4 highly expressed in the fat-tail will play an important role for tail fat deposition. Our study might provide a novel view to understanding fat accumulation in special parts of the body in sheep and other animals.


Subject(s)
Adipose Tissue/metabolism , Adipose Tissue/physiology , Sheep, Domestic/metabolism , Adipocytes/metabolism , Animals , China , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Fatty Acids/metabolism , Gene Expression/genetics , Lipid Metabolism/physiology , Peroxisomal Multifunctional Protein-2/genetics , Peroxisomal Multifunctional Protein-2/metabolism , Phenotype , Proteomics/methods , Sheep , Tail , Transcriptome/genetics
6.
Aging (Albany NY) ; 12(14): 14699-14717, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32678070

ABSTRACT

Steroidogenic enzymes are crucial in prostate cancer (PCa) progression. 17ß-Hydroxysteroid dehydrogenase type 4 (HSD17B4), encoded by HSD17B4, lacks catalytic capacity in androgen metabolism. Now the detailed role and molecular mechanism of PCa development are largely unknown. Here we showed that the expression of HSD17B4 was increased in PCa tissues compared to paired paratumor tissues. HSD17B4 knockdown in PCa cells significantly suppressed its proliferation, migration and invasion, while overexpressing HSD17B4 had opposite effects. Mechanistically, we found that the protein level of HSD17B4 was regulated by its acetylation at lysine 669(K669). Dihydroxytestosterone (DHT) treatment increased HSD17B4 acetylation and then promoted its degradation via chaperone-mediated autophagy (CMA). SIRT3 directly interacted with HSD17B4 to inhibit its acetylation and enhance its stability. In addition, we identified CREBBP as a regulator of the K669 acetylation and degradation of HSD17B4, affecting PC cell proliferation, migration and invasion. Notably, in PCa tissues and paired paratumor tissues, the level of HSD17B4 was negatively correlated with its K669 acetylation. Taken together, this study identified a novel role of HSD17B4 in PCa progression and suggested that HSD17B4 and its upstream regulators may be potential therapeutic targets for PCa intervention.


Subject(s)
Peroxisomal Multifunctional Protein-2/genetics , Peroxisomal Multifunctional Protein-2/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Acetylation , Animals , CREB-Binding Protein/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , Lysine/metabolism , Male , Mice , Mice, Knockout , Neoplasm Invasiveness/genetics , Sirtuin 3/genetics , Testosterone/metabolism , Xenograft Model Antitumor Assays
7.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(10): 1514-1524, 2019 10.
Article in English | MEDLINE | ID: mdl-31176039

ABSTRACT

The sphingolipid ceramide regulates beta-oxidation of medium and long chain fatty acids in mitochondria. It is not known whether it also regulates oxidation of very long chain fatty acids (VLCFAs) in peroxisomes. Using affinity chromatography, co-immunoprecipitation, and proximity ligation assays we discovered that ceramide interacts with Hsd17b4, an enzyme critical for peroxisomal VLCFA oxidation and docosahexaenoic acid (DHA) generation. Immunocytochemistry showed that Hsd17b4 is distributed to ceramide-enriched mitochondria-associated membranes (CEMAMs). Molecular docking and in vitro mutagenesis experiments showed that ceramide binds to the sterol carrier protein 2-like domain in Hsd17b4 adjacent to peroxisome targeting signal 1 (PTS1), the C-terminal signal for interaction with peroxisomal biogenesis factor 5 (Pex5), a peroxin mediating transport of Hsd17b4 into peroxisomes. Inhibition of ceramide biosynthesis induced translocation of Hsd17b4 from CEMAMs to peroxisomes, interaction of Hsd17b4 with Pex5, and upregulation of DHA. This data indicates a novel role of ceramide as a molecular switch regulating interaction of Hsd17b4 with Pex5 and peroxisomal function.


Subject(s)
Ceramides/metabolism , Peroxisomal Multifunctional Protein-2/metabolism , Peroxisome-Targeting Signal 1 Receptor/metabolism , Peroxisomes/metabolism , Animals , Cells, Cultured , Docosahexaenoic Acids/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Models, Molecular , Protein Interaction Maps , Protein Transport
9.
Chem Biol Interact ; 281: 69-80, 2018 Feb 01.
Article in English | MEDLINE | ID: mdl-29273565

ABSTRACT

Propiverine, a frequently-prescribed pharmaceutical for the treatment of symptoms associated with overactive bladder syndrome, provoked massive intranuclear and cytosolic protein inclusions in rat proximal tubule epithelium, primarily consisting of the peroxisomal targeting signal 1 (PTS1) containing protein d-amino acid oxidase (DAAO). As this type of nephropathy was also observed for other drugs, the aim was to determine whether propiverine interferes with trafficking and/or import of peroxisomal proteins. To elucidate this, DAAO- and propiverine-specific interaction partners from human HEK293 and rat WKPT cell lines and rat kidney and liver homogenate were determined using co-immunoprecipitation with subsequent nano-ESI-LC-MS/MS analyses. Corroboration of the role of DAAO- and/or propiverine-specific interaction partners in the drug-induced DAAO accumulation was sought via specific immunofluorescence staining of rat kidney sections from control and propiverine-treated rats. Above analyses demonstrated the interaction of propiverine with several protein classes, foremost peroxisomal proteins (DAAO, MFE2, HAOX2) and proteins of the protein quality control system, i.e. chaperones (HSP70 and DnaJ co-chaperones), proteases and proteasomal proteins (regulatory subunits of the 26S proteasome; Rpn1/2). The immunofluorescence analysis revealed mislocalization of many PTS1-proteins (DAAO, CAT, MFE2, ACOX1, EHHADH) in rat renal sections, strongly suggesting that propiverine primarily binds to PTS1 proteins resulting in the formation of PTS1 but not PTS2 or peroxisomal membrane protein (PMP) accumulations. Moreover, chaperones involved in peroxisomal trafficking (HSC70, DnaJB1) and peroxisomal biogenesis factor proteins (PEX3, PEX5, PEX7), also presented with distinct mislocalization patterns. Concomitantly, an increased number of peroxisomes was observed, suggestive of a compensatory mechanism for the presumably suboptimally functioning peroxisomes. Overall, the data presented suggested that propiverine interacts exclusively with DAAO or with a selected number of PTS1 proteins. The consequence of this interaction is the abrogated trafficking and peroxisomal import of PTS1 proteins concomitant with their nuclear and cytosolic accumulation due to inhibited degradation and imbalanced protein homeostasis.


Subject(s)
Amino Acid Oxidoreductases/metabolism , Benzilates/metabolism , Diabetic Nephropathies/etiology , 17-Hydroxysteroid Dehydrogenases/metabolism , Alcohol Oxidoreductases/metabolism , Amino Acid Oxidoreductases/chemistry , Amino Acid Oxidoreductases/genetics , Animals , Benzilates/chemistry , Benzilates/toxicity , Cell Line , Chromatography, High Pressure Liquid , HEK293 Cells , Humans , Immunoprecipitation , Kidney/metabolism , Kidney/pathology , Liver/metabolism , Microscopy, Confocal , Molecular Chaperones/metabolism , Peroxisomal Multifunctional Protein-2/metabolism , Peroxisome-Targeting Signal 1 Receptor/chemistry , Peroxisome-Targeting Signal 1 Receptor/metabolism , Protein Transport/drug effects , Rats , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
10.
Mol Imaging Biol ; 20(1): 94-102, 2018 02.
Article in English | MEDLINE | ID: mdl-28695372

ABSTRACT

PURPOSE: Activation of the innate immune system plays a significant role in pathologies of the central nervous system (CNS). In order to follow disease progression and evaluate effectiveness of potential treatments involved in neuroinflammation, it is important to track neuroinflammatory markers in vivo longitudinally. The translocator protein (TSPO) is used as a target to image neuroinflammation as its expression is upregulated in reactive glial cells during CNS pathologies. However, it remains unclear in which microglial phenotypes TSPO levels are upregulated, as microglia can display a plethora of activation states that can be protective or detrimental to the CNS. PROCEDURES: We assessed the levels of TSPO transcripts in cultured microglia that were polarized into pro- and anti-inflammatory states in vitro and in the brain of mice in which an anti-inflammatory environment was induced in vivo. In addition, we used a mouse model of peroxisomal multifunctional protein-2 (MFP2) deficiency that exhibits widespread neuroinflammation despite no neuronal loss and monitored TSPO expression by immunohistochemistry and by imaging using the TSPO radiotracer [18F]DPA-714. RESULTS: TSPO expression was selectively increased in so-called classically activated or M1 microglia but not in alternatively activated or M2 microglia in vitro. In agreement, TSPO transcript levels were not induced in an anti-inflammatory brain environment. We found that both transcript and protein levels of TSPO are significantly increased in the brain of Mfp2 -/- compared to those of the control mice and TSPO immunoreactivity colocalized predominantly with microglia in Mfp2 -/- brain. In vitro and ex vivo autoradiography in Mfp2 -/- mice using the TSPO radiotracer [18F]DPA-714 confirmed increased expression of TSPO. These data demonstrate that TSPO imaging reveals microgliosis in non-neurodegenerative brain pathologies. CONCLUSIONS: We show that induced TSPO expression marks a pro-inflammatory brain environment that is not necessarily accompanied by neuronal loss.


Subject(s)
Inflammation/pathology , Microglia/pathology , Nerve Degeneration/pathology , Receptors, GABA/genetics , Animals , Brain/pathology , Inflammation/genetics , Mice , Nerve Degeneration/genetics , Peroxisomal Multifunctional Protein-2/deficiency , Peroxisomal Multifunctional Protein-2/metabolism , Positron-Emission Tomography , Pyrazoles/metabolism , Pyrimidines/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, GABA/metabolism , Up-Regulation/genetics
11.
Org Biomol Chem ; 15(36): 7623-7629, 2017 Sep 20.
Article in English | MEDLINE | ID: mdl-28868548

ABSTRACT

Cytisine-linked isoflavonoids (CLIFs) inhibited PC-3 prostate and LS174T colon cancer cell proliferation by inhibiting a peroxisomal bifunctional enzyme. A pull-down assay using a biologically active, biotin-modified CLIF identified the target of these agents as the bifunctional peroxisomal enzyme, hydroxysteroid 17ß-dehydrogenase-4 (HSD17B4). Additional studies with truncated versions of HSD17B4 established that CLIFs specifically bind the C-terminus of HSD17B4 and selectively inhibited the enoyl CoA hydratase but not the d-3-hydroxyacyl CoA dehydrogenase activity. HSD17B4 was overexpressed in prostate and colon cancer tissues, knocking down HSD17B4 inhibited cancer cell proliferation, suggesting that HSD17B4 is a potential biomarker and drug target and that CLIFs are potential probes or therapeutic agents for these cancers.


Subject(s)
Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Isoflavones/pharmacology , Peroxisomal Multifunctional Protein-2/antagonists & inhibitors , Alkaloids/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Azocines/chemistry , Azocines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Isoflavones/chemical synthesis , Isoflavones/chemistry , Molecular Structure , Peroxisomal Multifunctional Protein-2/metabolism , Quinolizines/chemistry , Quinolizines/pharmacology , Structure-Activity Relationship
12.
BMC Med Genet ; 18(1): 91, 2017 08 23.
Article in English | MEDLINE | ID: mdl-28830375

ABSTRACT

BACKGROUND: Perrault syndrome is a rare multisystem disorder that manifests with sensorineural hearing loss in both sexes, primary ovarian insufficiency in females and neurological features. The syndrome is heterogeneous both genetically and phenotypically. CASE PRESENTATION: We reported a consanguineous family (two affected sisters) with Perrault syndrome. The proband had the characteristics of Perrault syndrome: ovarian dysgenesis, bilateral hearing loss and obvious neurological signs. Target genetic sequencing and triplet repeat primed PCR (TP-PCR) plus capillary electrophoresis was conducted to detect causative mutations in the proband. The detected variant was further confirmed in the proband and tested in other family members by Sanger sequencing. Both the proband and her sister were found homozygous for the novel variant HSD17B4 c.298G > T (p.A100S) with their parents heterozygous. Detected by western blot, the protein expression of HSD17B4 mutant was much lower than that of the wild type in SH-SY5Y cells transfected by HSD17B4 wild type or mutant plasmid, which indicated the pathogenicity of the HSD17B4 mutation. CONCLUSIONS: Our findings supported that HSD17B4 was one of the genes contributing to Perrault syndrome with the likely pathogenic variant c.298G > T (p.A100S). Special manifestations of cerebellar impairment were found in cases caused by HSD17B4 mutations. Besides, attention should be paid to distinguish Perrault syndrome from D-bifunctional protein deficiency and hereditary ataxia.


Subject(s)
Asian People/genetics , Gonadal Dysgenesis, 46,XX/genetics , Hearing Loss, Sensorineural/genetics , Homozygote , Mutation, Missense , Peroxisomal Multifunctional Protein-2/genetics , Adult , Cell Line , Female , Gene Expression Regulation , Genetic Testing , Gonadal Dysgenesis, 46,XX/diagnosis , Hearing Loss, Sensorineural/diagnosis , Heterozygote , Humans , Magnetic Resonance Imaging , Pedigree , Peroxisomal Multifunctional Protein-2/metabolism , Sequence Analysis, DNA
13.
Methods Mol Biol ; 1595: 329-342, 2017.
Article in English | MEDLINE | ID: mdl-28409475

ABSTRACT

The peroxisomal disorders (PDs) are a heterogeneous group of genetic diseases in man caused by an impairment in peroxisome biogenesis or one of the metabolic functions of peroxisomes. Thanks to the revolutionary technical developments in gene sequencing methods and their increased use in patient diagnosis, the field of genetic diseases in general and peroxisomal disorders in particular has dramatically changed in the last few years. Indeed, several novel peroxisomal disorders have been identified recently and in addition it has been realized that the phenotypic spectrum of patients affected by a PD keeps widening, which makes clinical recognition of peroxisomal patients increasingly difficult. Here, we describe these new developments and provide guidelines for the clinical and laboratory diagnosis of peroxisomal patients.


Subject(s)
Peroxisomal Disorders/diagnosis , Peroxisomal Disorders/genetics , Acyl-CoA Oxidase/deficiency , Acyl-CoA Oxidase/genetics , Genetic Testing , Humans , Peroxisomal Disorders/metabolism , Peroxisomal Multifunctional Protein-2/deficiency , Peroxisomal Multifunctional Protein-2/genetics , Peroxisomal Multifunctional Protein-2/metabolism , Peroxisomes/genetics , Peroxisomes/metabolism , Phenotype
14.
Autophagy ; 13(3): 538-553, 2017 Mar 04.
Article in English | MEDLINE | ID: mdl-28296597

ABSTRACT

Dysregulation of hormone metabolism is implicated in human breast cancer. 17ß-hydroxysteroid dehydrogenase type 4 (HSD17B4) catalyzes the conversion of estradiol (E2) to estrone (E1), and is associated with the pathogenesis and development of various cancers. Here we show that E1 upregulates HSD17B4 acetylation at lysine 669 (K669) and thereby promotes HSD17B4 degradation via chaperone-mediated autophagy (CMA), while a single mutation at K669 reverses the degradation and confers migratory and invasive properties to MCF7 cells upon E1 treatment. CREBBP and SIRT3 dynamically control K669 acetylation level of HSD17B4 in response to E1. More importantly, K669 acetylation is inversely correlated with HSD17B4 in human breast cancer tissues. Our study reveals a crosstalk between acetylation and CMA degradation in HSD17B4 regulation, and a critical role of the regulation in the malignant progression of breast cancer.


Subject(s)
Autophagy/drug effects , Estrone/pharmacology , Peroxisomal Multifunctional Protein-2/metabolism , Proteolysis/drug effects , Acetylation , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CREB-Binding Protein/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Lysine/metabolism , Models, Biological , Mutation/genetics , Neoplasm Invasiveness , Peroxisomal Multifunctional Protein-2/genetics , Sirtuin 3/metabolism , Substrate Specificity/drug effects
15.
Neurobiol Dis ; 94: 157-68, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27353294

ABSTRACT

The cerebellar pathologies in peroxisomal diseases underscore that these organelles are required for the normal development and maintenance of the cerebellum, but the mechanisms have not been resolved. Here we investigated the origins of the early-onset coordination impairment in a mouse model with neural selective deficiency of multifunctional protein-2, the central enzyme of peroxisomal ß-oxidation. At the age of 4weeks, Nestin-Mfp2(-/-) mice showed impaired motor learning on the accelerating rotarod and underperformed on the balance beam test. The gross morphology of the cerebellum and Purkinje cell arborization were normal. However, electrophysiology revealed a reduced Purkinje cell firing rate, a decreased excitability and an increased membrane capacitance. The distribution of climbing and parallel fiber synapses on Purkinje cells was immature and was accompanied by an increased spine length. Despite normal myelination, Purkinje cell axon degeneration was evident from the occurrence of axonal swellings containing accumulated organelles. In conclusion, the electrical activity, axonal integrity and wiring of Purkinje cells are exquisitely dependent on intact peroxisomal ß-oxidation in neural cells.


Subject(s)
Cerebellum/metabolism , Peroxisomal Multifunctional Protein-2/metabolism , Purkinje Cells/metabolism , Synapses/physiology , Animals , Axons/metabolism , Cerebellar Ataxia/metabolism , Mice, Knockout , Peroxisomal Multifunctional Protein-2/deficiency
16.
Sci Rep ; 5: 15136, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26456697

ABSTRACT

SQAP is a novel and promising anticancer agent that was obtained by structural modifications from a natural compound. SQAP inhibits angiogenesis in vivo resulting in increased hypoxia and reduced tumor volume. In this study, the mechanism by which SQAP modifies the tumor microenvironment was revealed through the application of a T7 phage display screening. This approach identified five SQAP-binding proteins including sterol carrier protein 2, multifunctional enzyme type 2, proteasomal ubiquitin receptor, UV excision repair protein and focal adhesion kinase (FAK). All the interactions were confirmed by surface plasmon resonance analysis. Since FAK plays an important role in cell turnover and angiogenesis, the influence of SQAP on FAK was the principal goal of this study. SQAP decreased FAK phosphorylation and cell migration in human umbilical vein endothelial cells and A549 cancer cells. These findings suggest that inhibition of FAK phosphorylation works as the mechanism for the anti-angiogenesis activity of SQAP.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Small Cell/drug therapy , Focal Adhesion Kinase 1/antagonists & inhibitors , Glycolipids/pharmacology , Lung Neoplasms/drug therapy , Amino Acid Sequence , Animals , Antineoplastic Agents/chemical synthesis , Binding Sites , Carcinoma, Small Cell/enzymology , Carcinoma, Small Cell/genetics , Carcinoma, Small Cell/pathology , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Focal Adhesion Kinase 1/chemistry , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Glycolipids/chemical synthesis , Human Umbilical Vein Endothelial Cells , Humans , Intracellular Signaling Peptides and Proteins , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Nude , Molecular Docking Simulation , Molecular Sequence Data , Peptide Library , Peroxisomal Multifunctional Protein-2/chemistry , Peroxisomal Multifunctional Protein-2/genetics , Peroxisomal Multifunctional Protein-2/metabolism , Phosphorylation/drug effects , Protein Binding , Xenograft Model Antitumor Assays
17.
PLoS One ; 10(6): e0128749, 2015.
Article in English | MEDLINE | ID: mdl-26046837

ABSTRACT

The discrete effects of obesity on infertility in females remain undefined to date. To investigate obesity-induced ovarian dysfunction, we characterized metabolic parameters, steroidogenesis, and folliculogenesis in obese and lean female Ossabaw mini-pigs. Nineteen nulliparous, sexually mature female Ossabaw pigs were fed a high fat/cholesterol/fructose diet (n=10) or a control diet (n=9) for eight months. After a three-month diet-induction period, pigs remained on their respective diets and had ovarian ultrasound and blood collection conducted during a five-month study period after which ovaries were collected for histology, cell culture, and gene transcript level analysis. Blood was assayed for steroid and protein hormones. Obese pigs developed abdominal obesity and metabolic syndrome, including hyperglycemia, hypertension, insulin resistance and dyslipidemia. Obese pigs had elongated estrous cycles and hyperandrogenemia with decreased LH, increased FSH and luteal phase progesterone, and increased numbers of medium, ovulatory, and cystic follicles. Theca cells of obese, compared to control, pigs displayed androstenedione hypersecretion in response to in vitro treatment with LH, and up-regulated 3-beta-hydroxysteroid dehydrogenase 1 and 17-beta-hydroxysteroid dehydrogenase 4 transcript levels in response to in vitro treatment with LH or LH + insulin. Granulosa cells of obese pigs had increased 3-beta-hydroxysteroid dehydrogenase 1 transcript levels. In summary, obese Ossabaw pigs have increased transcript levels and function of ovarian enzymes in the delta 4 steroidogenic pathway. Alterations in LH, FSH, and progesterone, coupled with theca cell dysfunction, contribute to the hyperandrogenemia and disrupted folliculogenesis patterns observed in obese pigs. The obese Ossabaw mini-pig is a useful animal model in which to study the effects of obesity and metabolic syndrome on ovarian function and steroidogenesis. Ultimately, this animal model may be useful toward the development of therapies to improve fertility in obese and/or hyperandrogenemic females or in which to examine the effects of obesity on the maternal-fetal environment and offspring health.


Subject(s)
Androstenedione/blood , Follicle Stimulating Hormone/blood , Infertility, Female/metabolism , Luteinizing Hormone/blood , Metabolic Syndrome/metabolism , Obesity/metabolism , Progesterone/blood , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Animals , Diet, High-Fat/adverse effects , Estrous Cycle , Female , Follicle Stimulating Hormone/pharmacology , Gene Expression , Granulosa Cells/metabolism , Granulosa Cells/pathology , Humans , Infertility, Female/etiology , Infertility, Female/genetics , Infertility, Female/pathology , Insulin/pharmacology , Luteinizing Hormone/pharmacology , Metabolic Syndrome/etiology , Metabolic Syndrome/genetics , Metabolic Syndrome/pathology , Obesity/etiology , Obesity/genetics , Obesity/pathology , Peroxisomal Multifunctional Protein-2/genetics , Peroxisomal Multifunctional Protein-2/metabolism , Primary Cell Culture , RNA, Messenger/agonists , RNA, Messenger/genetics , RNA, Messenger/metabolism , Swine , Swine, Miniature , Theca Cells/drug effects , Theca Cells/metabolism , Theca Cells/pathology
18.
Lipids ; 50(9): 839-46, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25964052

ABSTRACT

We have previously reported that phosphoinositide 3-kinase p110δ knockout (p110δ KO) diminished the adhesion of leukocytes to capillary venules and suppressed the peritoneal infiltration of leukocytes, both functions that play important roles in atherosclerosis. Therefore, we hypothesized that p110δ deficiency might be protective against atherosclerosis. Apolipoprotein E knockout (ApoE KO) mice were crossed with p110δ KO mice to generate homozygous double knockout mice (ApoE/p110δ DKO). The present study showed that ApoE/p110δ DKO mice fed with a high cholesterol diet (HCD) demonstrated less peritoneal infiltration of leukocytes and monocytes compared with ApoE KO mice after intraperitoneal injection of thioglycollate, an inducer of acute peritoneal inflammation. Unexpectedly, atherosclerosis in the aortic root and in the entire aorta was similar between the ApoE/p110δ DKO and ApoE KO groups. No difference in Mac-3 expression, indicative of macrophage infiltration, was found between the two groups. Further analysis showed that ApoE KO mice chronically fed with HCD had increased levels of total cholesterol, low-density lipoprotein in the blood and counts and percentages of circulating monocytes compared with ApoE KO mice fed with a normal diet. Consistently, the deficiency of p110δ affected neither the counts nor the percentages of monocytes nor the lipid profiles in the blood. The results suggested that p110δ plays an important role in acute but not in chronic inflammation, the latter being included in the early characteristics of atherosclerosis, which might explain the finding that p110δ deficiency fails to inhibit early atherosclerosis.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/metabolism , Peritoneum/pathology , Phosphatidylinositol 3-Kinases/metabolism , Animals , Apolipoproteins E/metabolism , Atherosclerosis/immunology , Cholesterol/blood , Class I Phosphatidylinositol 3-Kinases , Diet, High-Fat , Inflammation/metabolism , Leukocytes/immunology , Male , Mice , Mice, Knockout , Monocytes/immunology , Peroxisomal Multifunctional Protein-2/metabolism , Phosphatidylinositol 3-Kinases/genetics , Thioglycolates/metabolism
19.
Bioorg Med Chem Lett ; 25(21): 4976-4979, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-25804720

ABSTRACT

ß-Sitosterol is the most abundant plant sterol in the human diet. It is also the major component of several traditional medicines, including saw palmetto and devil's claw. Although ß-sitosterol is effective against enlarged prostate in human clinical trials and has anti-cancer and anti-inflammatory activities, the mechanisms of action are poorly understood. Here, we report the identification of two new binding proteins for ß-sitosterol that may underlie its beneficial effects.


Subject(s)
Peroxisomal Multifunctional Protein-2/metabolism , Sitosterols/metabolism , Sitosterols/pharmacology , Synaptotagmin I/metabolism , Animals , Binding Sites , Humans , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Mice , Molecular Conformation , Peroxisomal Multifunctional Protein-2/chemistry , Protein Binding , Sitosterols/chemistry , Synaptotagmin I/chemistry
20.
Mol Cell Endocrinol ; 401: 1-11, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25448063

ABSTRACT

Hepatocellular carcinoma (HCC) arises in a setting of chronic inflammation induced by inflammatory cytokines, such as nuclear factor-kappaB (NF-κB). HCC is a male-predominant cancer that can be attenuated by estradiol (E2) in vitro and in vivo. Although 17ß-hydroxysteroid dehydrogenase 4 (HSD17B4) has been implicated as an estradiol-inactivating enzyme, and its promoter sequence contains two putative NF-κB elements: it is currently unknown whether HSD17B4 is the link between inflammation, estradiol and proliferation in hepatoma cells. In this study, HepG2 cells were used to investigate the role of HSD17B4 in the proliferation of liver cancer cells treated with the NF-κB activator, tumor necrosis factor-alpha (TNF-α), with the inhibitor of NF-κB activation, pyrrolidinedithiocarbamate (PDTC), or with a related specific siRNA. We demonstrated that the human HSD17B4 gene is a target for NF-κB activation in inflammation-stimulated HepG2 cells. HSD17B4 is up-regulated via the binding of activated NF-κB to the distal NF-κB-responsive element via TNF-α stimulation, which then promotes cell proliferation by decreasing the levels of E2 and enhancing the expression of interleukin 6 (IL-6), cyclin D1 and proliferating cell nuclear antigen (PCAN). These results from HepG2 cells are consistent with the observation that HSD17B4 is highly expressed and activated NF-κB is highly co-localized with the NF-κB-responsive element of HSD17B4 in liver tumor tissues from HCC patients. Our findings indicate for the first time that HSD17B4 plays an important role in aggravated HCC progression and provides a novel therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Estradiol/metabolism , Liver Neoplasms/pathology , NF-kappa B/metabolism , Peroxisomal Multifunctional Protein-2/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Proliferation/drug effects , Cyclin D1/metabolism , Hep G2 Cells , Humans , Interleukin-6/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Peroxisomal Multifunctional Protein-2/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Proline/analogs & derivatives , Proline/pharmacology , Promoter Regions, Genetic , Thiocarbamates/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...