Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Tissue Res ; 357(1): 267-78, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24770931

ABSTRACT

Acyl-CoA synthetase 5 (ACSL5), a mitochondrially localized enzyme, catalyzes the synthesis of long-chain fatty acid thioesters and is physiologically involved in pro-apoptotic sensing of enterocytes. The aim of the present study is to identify an ACSL5-dependent regulation of mitochondrially expressed proteins and the characterization of related pathways in normal and diseased human intestinal mucosa. Proteomics of isolated mitochondria from ACSL5 transfectants and CaCo2 controls were performed. ACSL5-dependent protein synthesis was verified with quantitative reverse transcription plus the polymerase chain reaction, Western blotting, short-interfering-RNA-mediated gene silencing and additional cell culture experiments. Lipid changes were analyzed with tandem mass spectrometry. ACSL5-related pathways were characterized in normal mucosa and sporadic adenocarcinomas of the human intestine. In CaCo2 cells transfected with ACSL5, mortalin (HSPA9) was about two-fold increased in mitochondria, whereas cytoplasmic mortalin levels were unchanged. Disturbance of acyl-CoA/sphingolipid metabolism, induced by ACSL5 over-expression, was characterized as crucial. ACSL5-related over-expression of mitochondrial mortalin was found in HEK293 and Lovo (wild-type TP53 [tumor protein p53]) and CaCo2 (p53-negative; TP53 mutated) cells but not in Colo320DM cells (mutated TP53). In normal human intestinal mucosa, an increasing gradient of both ACSL5 and mortalin from bottom to top was observed, whereas p53 (wild-type TP53) decreased. In sporadic intestinal adenocarcinomas with strong p53 immunostaining (mutated TP53), ACSL5-related mortalin expression was heterogeneous. ACSL5-induced mitochondrial mortalin expression is assumed to be a stress response to ACSL5-related changes in lipid metabolism and is regulated by the TP53 status. Uncoupling of ACSL5 and mitochondrial mortalin by mutated TP53 could be important in colorectal carcinogenesis.


Subject(s)
Coenzyme A Ligases/biosynthesis , Colorectal Neoplasms/metabolism , Enterocytes/metabolism , HSP70 Heat-Shock Proteins/genetics , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Adult , Aged , Caco-2 Cells , Cloning, Molecular , Coenzyme A Ligases/genetics , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Enterocytes/enzymology , Enterocytes/pathology , Female , Humans , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism , Male , Middle Aged , Mitochondria/enzymology , Transfection
2.
Histol Histopathol ; 28(3): 353-64, 2013 03.
Article in English | MEDLINE | ID: mdl-23348389

ABSTRACT

Metabolic components like fatty acids and acyl-Coenzyme A (acyl-CoA) thioesters have been implicated in the pathogenesis of various tumours. The activation of fatty acids to acyl-CoAs is catalysed by long chain acyl-CoA synthetases (ACSLs), and impairment of ACSL expression levels has been associated with tumourigenesis and progression. Since ACSLs have never been investigated in bladder tissues, the study aims to characterize ACSL expression and acyl-CoA synthesis in normal and neoplastic bladder tissues, as well as cell lines. ACSL isoforms 1, 3, 4 and 5 and synthesis of acyl-CoAs were analysed using qRT-PCR, western blot analysis, immunohistochemistry and lipid mass spectrometry. In normal urothelium, expression of ACSL1, 3, 4 and 5, with highest levels of ACSL isoform 5 was found. However, ACSL5 expression was reduced in corresponding neoplastic tissues and urothelial cell lines depending on the grade of cellular differentiation. Anti-ACSL5 immunostainings showed expression in normal urothelium and a gradual loss of ACSL5 protein via pre-invasive lesions to invasive carcinomas. High expression of ACSL5 correlated with increased α-galactosidase activity and positive Uroplakin III staining in tumours. In contrast, synthesis of acyl-CoAs was enhanced in neoplastic bladder tissues compared to normal urothelium, and reflected an increase with respect to cellular differentiation. These results confirm an expression of ACSLs, especially isoform 5, in human urothelium, prove enzymatic/lipidomic changes in bladder cancer tissues, and suggest an involvement of ACSL5 in cellular maturation and/or senescence with possible effects onto induction of tumour formation or progression. Further work may identify responsible pathway alterations, and attempting to re-balance the metabolic equilibrium of the urothelium may offer a further opportunity for tumour treatment and prevention.


Subject(s)
Carcinoma, Transitional Cell/enzymology , Coenzyme A Ligases/metabolism , Urinary Bladder Neoplasms/enzymology , Urinary Bladder/enzymology , Urothelium/enzymology , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/pathology , Cell Line, Transformed , Cell Line, Tumor , Coenzyme A Ligases/genetics , Cystectomy , Gene Expression , Humans , Isoenzymes , Urinary Bladder/pathology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Urothelium/pathology
3.
Histopathology ; 60(4): 561-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22296301

ABSTRACT

AIMS: Deleted in malignant brain tumours 1 (DMBT1; gp340) is a secreted glycoprotein which is found in the surface lining epithelia of human small and large intestine. DMBT1 is suggested to play a role in enterocyte differentiation and surface protection from intestinal bacteria. The aim of this study was to elucidate DMBT1 expression in bacteria-related active intestinal inflammation such as appendicitis. METHODS AND RESULTS: mRNA and protein levels of DMBT1 were analysed in surgical resections of 50 appendices (active inflammation: n = 25). In non-actively inflamed appendices, inter-individual differences in basal DMBT1 levels of enterocytes and some non-epithelial cells were found. In active appendicitis, enterocytic DMBT1 mRNA expression was increased approximately fivefold, which was paralleled by a corresponding increase of cytoplasmic and secreted DMBT1 protein levels. Increased DMBT1 expression was predominant in enterocytes adjacent to erosive lesions or ulcers. CONCLUSIONS: Our data demonstrate that bacteria-related active inflammation results in a sharp increase of DMBT1 levels in enterocytes. These findings substantiate the view that DMBT1 is of functional relevance for host defence and modulation of the course of intestinal bacteria-related inflammatory responses.


Subject(s)
Appendicitis/metabolism , Appendix/metabolism , Enterocytes/metabolism , Receptors, Cell Surface/metabolism , Adolescent , Adult , Aged , Appendicitis/genetics , Appendicitis/pathology , Appendix/pathology , Calcium-Binding Proteins , Child , DNA-Binding Proteins , Enterocytes/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Male , Middle Aged , Receptors, Cell Surface/genetics , Tumor Suppressor Proteins
4.
PLoS Genet ; 8(2): e1002488, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22319455

ABSTRACT

Overexpression or mutation of α-Synuclein is associated with protein aggregation and interferes with a number of cellular processes, including mitochondrial integrity and function. We used a whole-genome screen in the fruit fly Drosophila melanogaster to search for novel genetic modifiers of human [A53T]α-Synuclein-induced neurotoxicity. Decreased expression of the mitochondrial chaperone protein tumor necrosis factor receptor associated protein-1 (TRAP1) was found to enhance age-dependent loss of fly head dopamine (DA) and DA neuron number resulting from [A53T]α-Synuclein expression. In addition, decreased TRAP1 expression in [A53T]α-Synuclein-expressing flies resulted in enhanced loss of climbing ability and sensitivity to oxidative stress. Overexpression of human TRAP1 was able to rescue these phenotypes. Similarly, human TRAP1 overexpression in rat primary cortical neurons rescued [A53T]α-Synuclein-induced sensitivity to rotenone treatment. In human (non)neuronal cell lines, small interfering RNA directed against TRAP1 enhanced [A53T]α-Synuclein-induced sensitivity to oxidative stress treatment. [A53T]α-Synuclein directly interfered with mitochondrial function, as its expression reduced Complex I activity in HEK293 cells. These effects were blocked by TRAP1 overexpression. Moreover, TRAP1 was able to prevent alteration in mitochondrial morphology caused by [A53T]α-Synuclein overexpression in human SH-SY5Y cells. These results indicate that [A53T]α-Synuclein toxicity is intimately connected to mitochondrial dysfunction and that toxicity reduction in fly and rat primary neurons and human cell lines can be achieved using overexpression of the mitochondrial chaperone TRAP1. Interestingly, TRAP1 has previously been shown to be phosphorylated by the serine/threonine kinase PINK1, thus providing a potential link of PINK1 via TRAP1 to α-Synuclein.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons/metabolism , Drosophila melanogaster/genetics , HSP90 Heat-Shock Proteins/metabolism , Mitochondria/metabolism , Molecular Chaperones/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Adenosine Triphosphate/biosynthesis , Adenosine Triphosphate/genetics , Animals , Cell Survival/genetics , Dopamine/physiology , Dopaminergic Neurons/drug effects , Gene Expression Regulation/drug effects , Gene Silencing , HEK293 Cells , HSP90 Heat-Shock Proteins/genetics , Humans , Membrane Potential, Mitochondrial , Mitochondria/genetics , Molecular Chaperones/genetics , Mutation , Oxidative Stress , PC12 Cells , Parkinson Disease/genetics , Parkinson Disease/metabolism , RNA, Small Interfering , Rats , Rotenone/pharmacology , alpha-Synuclein/toxicity
5.
World J Gastroenterol ; 17(44): 4883-9, 2011 Nov 28.
Article in English | MEDLINE | ID: mdl-22171129

ABSTRACT

AIM: To investigate whether human acyl-CoA synthetase 5 (ACSL5) is sensitive to the ACSL inhibitor triacsin C. METHODS: The ACSL isoforms ACSL1 and ACSL5 from rat as well as human ACSL5 were cloned and recombinantly expressed as 6xHis-tagged enzymes. Ni(2+)-affinity purified recombinant enzymes were assayed at pH 7.5 or pH 9.5 in the presence or absence of triacsin C. In addition, ACSL5 transfected CaCo2 cells and intestinal human mucosa were monitored. ACSL5 expression in cellular systems was verified using Western blot and immunofluorescence. The ACSL assay mix included TrisHCl (pH 7.4), ATP, CoA, EDTA, DTT, MgCl(2), [9,10-(3)H] palmitic acid, and triton X-100. The 200 µL reaction was initiated with the addition of solubilized, purified recombinant proteins or cellular lysates. Reactions were terminated after 10, 30 or 60 min of incubation with Doles medium. RESULTS: Expression of soluble recombinant ACSL proteins was found after incubation with isopropyl beta-D-1-thiogalactopyranoside and after ultracentrifugation these were further purified to near homogeneity with Ni(2+)-affinity chromatography. Triacsin C selectively and strongly inhibited recombinant human ACSL5 protein at pH 7.5 and pH 9.5, as well as recombinant rat ACSL1 (sensitive control), but not recombinant rat ACSL5 (insensitive control). The IC50 for human ACSL5 was about 10 µmol/L. The inhibitory triacsin C effect was similar for different incubation times (10, 30 and 60 min) and was not modified by the N- or C-terminal location of the 6xHis-tag. In order to evaluate ACSL5 sensitivity to triacsin C in a cellular environment, stable human ACSL5 CaCo2 transfectants and mechanically dissected normal human intestinal mucosa with high physiological expression of ACSL5 were analyzed. In both models, ACSL5 peak activity was found at pH 7.5 and pH 9.5, corresponding to the properties of recombinant human ACSL5 protein. In the presence of triacsin C (25 µmol/L), total ACSL activity was dramatically diminished in human ACSL5 transfectants as well as in ACSL5-rich human intestinal mucosa. CONCLUSION: The data strongly indicate that human ACSL5 is sensitive to triacsin C and does not compensate for other triacsin C-sensitive ACSL isoforms.


Subject(s)
Coenzyme A Ligases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/enzymology , Isoenzymes/antagonists & inhibitors , Mitochondrial Proteins/antagonists & inhibitors , Triazenes/pharmacology , Animals , Cell Line , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Humans , Inhibitory Concentration 50 , Isoenzymes/genetics , Isoenzymes/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
6.
Biochim Biophys Acta ; 1801(9): 1025-35, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20470896

ABSTRACT

In the pathogenesis of nonalcoholic fatty liver disease, accumulation of lipids in hepatocytes and hepatocyte apoptosis are strongly implicated in disease progression from the potentially reversible condition of steatosis to severe acute and chronic liver injury. Acyl-CoA synthetase 5, a member of the ACSL gene family that catalyzes the activation of long-chain fatty acids for lipid biosynthesis, is the only ACSL isoform that is both, located on mitochondria and functionally involved in enterocyte apoptosis. In this study, the regulation of human ACSL5 in hepatocellular fatty acid degeneration and its involvement in hepatocyte apoptosis was investigated using models of in vitro and in vivo steatosis as well as plasmid-mediated stable gene transfer and RNAi-mediated gene silencing. ACSL5 mRNA and protein were strongly increased by uptake of dietary derived fatty acids in primary human hepatocytes, HepG2 cells and human steatotic liver. Over-expression of ACSL5 decreased HepG2 cell viability and increased susceptibility to TRAIL- and TNFalpha-, but not FAS- induced apoptosis, whereas knock down of ACSL5 reduced apoptosis susceptibility. High ACSL5 activity resulted in enhanced caspase-3/7 activity, but was not accompanied by up-regulation of death receptors, DR4, DR5 or TNF-R1. This study gives evidence that hepatocyte steatosis is associated with ACSL5 up-regulation resulting in increased susceptibility to hepatic cell death. We propose that ACSL5 could play a role in promoting fatty acid-induced lipoapoptosis in hepatocytes as important mechanism in fatty liver-related disorders.


Subject(s)
Apoptosis , Coenzyme A Ligases/metabolism , Fatty Liver/pathology , Hepatocytes/pathology , Lipids/pharmacology , Liver/pathology , Blotting, Western , Cell Proliferation , Coenzyme A Ligases/antagonists & inhibitors , Coenzyme A Ligases/genetics , Enzyme-Linked Immunosorbent Assay , Fatty Liver/enzymology , Fatty Liver/genetics , Fluorescent Antibody Technique , Hep G2 Cells , Hepatocytes/enzymology , Humans , Immunoenzyme Techniques , Liver/enzymology , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
7.
World J Gastroenterol ; 16(15): 1820-7, 2010 Apr 21.
Article in English | MEDLINE | ID: mdl-20397257

ABSTRACT

In the modifier concept of intestinal carcinogenesis, lipids have been established as important variables and one focus is given to long-chain fatty acids. Increased consumption of long-chain fatty acids is in discussion to modify the development of colorectal carcinoma in humans. Saturated long-chain fatty acids, in particular, are assumed to promote carcinogenesis, whereas polyunsaturated forms are likely to act in the opposite way. At present, the molecular mechanisms behind these effects are not well understood. Recently, it has been demonstrated by lipidomics and associated molecular techniques, that activation and metabolic channeling of long-chain fatty acids are important mechanisms to modify colorectal carcinogenesis. In this Editorial, an overview about the present concept of long-chain fatty acids and its derivatives in colorectal carcinogenesis as well as technical algorithms in lipid analysis is given.


Subject(s)
Colorectal Neoplasms/metabolism , Lipids/chemistry , Acyl Coenzyme A/metabolism , Apoptosis , Carcinogens , Dietary Fats , Disease Progression , Fatty Acids/metabolism , Humans , Models, Biological , Proteomics/methods
8.
BMC Gastroenterol ; 10: 27, 2010 Mar 07.
Article in English | MEDLINE | ID: mdl-20205943

ABSTRACT

BACKGROUND: Maturation of enterocytes along the small intestinal crypt-villus axis is associated with significant changes in gene expression profiles. fls485 coding a putative chaperone protein has been recently suggested as a gene involved in this process. The aim of the present study was to analyze fls485 expression in human small intestinal mucosa. METHODS: fls485 expression in purified normal or intestinal mucosa affected with celiac disease was investigated with a molecular approach including qRT-PCR, Western blotting, and expression strategies. Molecular data were corroborated with several in situ techniques and usage of newly synthesized mouse monoclonal antibodies. RESULTS: fls485 mRNA expression was preferentially found in enterocytes and chromaffine cells of human intestinal mucosa as well as in several cell lines including Rko, Lovo, and CaCo2 cells. Western blot analysis with our new anti-fls485 antibodies revealed at least two fls485 proteins. In a functional CaCo2 model, an increase in fls485 expression was paralleled by cellular maturation stage. Immunohistochemistry demonstrated fls485 as a cytosolic protein with a slightly increasing expression gradient along the crypt-villus axis which was impaired in celiac disease Marsh IIIa-c. CONCLUSIONS: Expression and synthesis of fls485 are found in surface lining epithelia of normal human intestinal mucosa and deriving epithelial cell lines. An interdependence of enterocyte differentiation along the crypt-villus axis and fls485 chaperone activity might be possible.


Subject(s)
Celiac Disease/genetics , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Molecular Chaperones/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Celiac Disease/metabolism , Cell Line , Child , Chromaffin System/metabolism , Enterocytes/metabolism , Female , Gene Expression Regulation , Humans , Male , Mice , Middle Aged , Open Reading Frames , RNA, Messenger/analysis , Reference Values , Young Adult
9.
World J Gastrointest Pathophysiol ; 1(5): 147-53, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21607156

ABSTRACT

Free fatty acids are essential dietary components and recognized as important molecules in the maintenance of cellular homeostasis. In the last decade, the molecular pathways for free fatty acid sensing in the gastrointestinal tract have been further elucidated by molecular identification and functional characterization of fatty acid binding receptors. These sensing molecules belong to the family of G protein-coupled receptors. In the intestine, four important receptors have been described so far. They differ in molecular structure, ligand specificity, expression pattern, and functional properties. In this review, an overview of intestinal fatty acid binding receptors and their role in intestinal physiology and pathophysiology is given.

SELECTION OF CITATIONS
SEARCH DETAIL
...