Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Pathobiology ; 74(3): 145-58, 2007.
Article in English | MEDLINE | ID: mdl-17643060

ABSTRACT

Defective protein folding is responsible for many diseases. Although these diseases seem to be quite diverse at the first glance, there is evidence for common pathogenetic principles. The basis of the pathological changes is the cell's inability to prevent protein misfolding, to revert misfolded proteins to normal or to eliminate misfolded proteins by degradation. This could result in deposition of potentially cytotoxic protein aggregates (protein aggregation diseases). Chronic degenerative diseases of the central nervous system (e.g. Alzheimer's and Parkinson's disease), the amyloidoses, but also chronic liver diseases, for example alcoholic and nonalcoholic steatohepatitis, belong to this category of disorders. This review highlights general pathogenic principles of protein aggregation diseases based on immunohistochemical and biochemical studies as well as observations in a mouse model for protein aggregation in the context of alcoholic and nonalcoholic steatohepatitis. The cellular defense mechanisms involved in protein quality control as well as the pathogenesis of protein aggregation diseases will be discussed.


Subject(s)
Molecular Chaperones/metabolism , Neurodegenerative Diseases/metabolism , Protein Folding , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloidosis/metabolism , Amyloidosis/pathology , Animals , Brain/metabolism , Brain/pathology , Chronic Disease , Disease Models, Animal , Fatty Liver, Alcoholic/metabolism , Fatty Liver, Alcoholic/pathology , Humans , Mice , Molecular Chaperones/chemistry , Neurodegenerative Diseases/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Conformation
2.
J Pathol ; 208(5): 653-61, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16477590

ABSTRACT

Mallory bodies (MBs) and intracellular hyaline bodies (IHBs) are cytoplasmic hepatocellular inclusions that consist of aggregated proteins. MBs are characteristically associated with alcoholic and non-alcoholic steatohepatitis, but may also be found in chronic cholestatic and metabolic (eg copper intoxication) diseases and hepatocellular neoplasms, particularly hepatocellular carcinomas. IHBs have hitherto only been described in hepatocellular carcinoma cells. In the present study hepatocellular carcinomas (HCCs) and a case of idiopathic copper toxicosis were evaluated with respect to the presence and mutual relationship of MBs and IHBs. IHBs alone were present in 8.6%, MBs alone in 16.1% and both types of inclusion in 7.5% of HCCs. It is shown that IHBs may also occur in non-neoplastic hepatocytes in association with idiopathic copper toxicosis, together with MBs. In HCCs and idiopathic copper toxicosis, MBs and IHBs may be present within the same cell. Moreover, hybrid inclusions holding an intermediate position between MBs and IHBs regarding light microscopy, ultrastructure and composition exist. MBs and IHBs contain p62, a stress-inducible adapter protein, as the major constituent. In MBs p62 is associated with keratins, whereas classical IHBs lack keratins. Light microscopic, electron microscopic and immunohistochemical data suggest a close pathogenetic relationship between MBs and IHBs. Both types of inclusion are the result of over-expression and accumulation of the stress protein p62. If p62 is induced alone, or at least prevails, IHBs may arise by aggregation. However, if abnormal keratins are present in addition to p62, p62 associates and co-aggregates with keratins, finally leading to classical MBs.


Subject(s)
Carcinoma, Hepatocellular/ultrastructure , Hepatocytes/ultrastructure , Inclusion Bodies/ultrastructure , Liver Neoplasms/ultrastructure , Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Hepatocellular/metabolism , Chemical and Drug Induced Liver Injury , Copper/poisoning , Hepatocytes/metabolism , Humans , Hyalin , Inclusion Bodies/metabolism , Keratins/metabolism , Liver Diseases/pathology , Liver Neoplasms/metabolism , Microscopy, Electron , Microscopy, Immunoelectron , Neoplasm Proteins/metabolism , Sequestosome-1 Protein
3.
Gut ; 55(5): 719-27, 2006 May.
Article in English | MEDLINE | ID: mdl-16322111

ABSTRACT

BACKGROUND: Bile acid induced apoptosis in hepatocytes can be antagonised by nuclear factor kappaB (NFkappaB) dependent survival pathways. Sulfasalazine modulates NFkappaB in different cell types. We aimed to determine the effects of sulfasalazine and its metabolites sulfapyridine and 5-aminosalicylic acid (5-ASA) on bile acid induced apoptosis in hepatocytes. METHODS: Apoptosis was determined by caspase assays and immunoblotting, NFkappaB activation by electrophoretic mobility shift assay and reporter gene assays, generation of reactive oxygen species (ROS) fluorometrically, bile secretion gravimetrically, and bile acid uptake radiochemically and by gas chromatography in HepG2-Ntcp cells and isolated perfused rat livers. RESULTS: Glycochenodeoxycholic acid (GCDCA 75 micromol/l) induced apoptosis was reduced by sulfasalazine dose dependently (1-1000 micromol/l) in HepG2-Ntcp cells whereas its metabolites 5-ASA and sulfapyridine had no effect. Sulfasalazine significantly reduced GCDCA induced activation of caspases 9 and 3. In addition, sulfasalazine activated NFkappaB and decreased GCDCA induced generation of ROS. Bile acid uptake was competitively inhibited by sulfasalazine. In perfused rat livers, GCDCA (25 micromol/l) induced liver injury and extensive hepatocyte apoptosis were significantly reduced by simultaneous administration of 100 micromol/l sulfasalazine: lactate dehydrogenase and glutamate-pyruvate transaminase activities were reduced by 82% and 87%, respectively, and apoptotic hepatocytes were observed only occasionally. GCDCA uptake was reduced by 45 (5)% when sulfasalazine was coadministered. However, when 50% of GCDCA (12.5 micromol/l) was administered alone, marked hepatocyte apoptosis and liver injury were again observed, questioning the impact of reduced GCDCA uptake for the antiapoptotic effect of sulfasalazine. CONCLUSION: Sulfasalazine is a potent inhibitor of GCDCA induced hepatocyte apoptosis in vitro and in the intact liver.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Glycochenodeoxycholic Acid/pharmacology , Hepatocytes/pathology , Sulfasalazine/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Apoptosis/drug effects , Caspase 3 , Caspase 9 , Caspases/metabolism , Cell Line, Tumor , Depression, Chemical , Dose-Response Relationship, Drug , Fluorescent Antibody Technique , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mesalamine/pharmacology , NF-kappa B/metabolism , Perfusion , Rats , Rats, Sprague-Dawley , Sulfapyridine/pharmacology , Sulfasalazine/metabolism
4.
Verh Dtsch Ges Pathol ; 89: 137-43, 2005.
Article in German | MEDLINE | ID: mdl-18035683

ABSTRACT

Morphologic criteria of steatohepatitis are steatosis, ballooning of hepatocytes, often but not constantly associated with Mallory bodies, pericellular fibrosis and inflammation. Liver cirrhosis follows in about 20-50%. With respect to etiology an alcoholic and non-alcoholic type can be distinguished, the latter being a characteristic hepatic lesion associated with the metabolic syndrome (type II diabetes, insulin resistance, obesity, dyslipidemia). Ballooning of hepatocytes as well as Mallory body formation are associated with a disturbance of the keratin intermediate filament cytoskeleton. Mallory bodies are protein aggregates consisting of keratin (particularly keratin 8), p62, a stress-induced adapter protein involved in signal transduction pathways, heat shock proteins, and ubiquitin. Oxidative stress is involved in Mallory body formation. Major sources of oxidative stress in alcoholic and non-alcoholic steatohepatitis are the microsomal biotransformation system (cytochrome P-450) and the mitochondria, together with an impaired antioxidant defense system. Oxidative stress leads to misfolding/unfolding, abnormal phosphorylation of keratins and disturbance of keratin 8: keratin 18 ratio, and thus interferes with intermediate filament assembly. Moreover, impairment of cellular defense against abnormal proteins, i. e. chaperone action and proteasomal degradation, leads to the accumulation of abnormal aggregation--prone keratins (particularly keratin 8) which after ubiquitination associate with the stress-induced ubiquitin-binding protein p62 to form Mallory bodies. Thus, Mallory body formation resembles an "off-folding" protein response of the amyloid type. These pathogenetic principles of the human disease are supported by immunohistochemical and gene expression studies in experimental animals and by transfection experiments in tissue culture cells.


Subject(s)
Fatty Liver, Alcoholic/pathology , Fatty Liver/pathology , Diagnosis, Differential , Fatty Liver/complications , Fatty Liver/physiopathology , Fatty Liver, Alcoholic/complications , Fatty Liver, Alcoholic/physiopathology , Humans , Inclusion Bodies/pathology , Keratins/analysis , Keratins/isolation & purification
5.
J Hepatol ; 34(5): 665-75, 2001 May.
Article in English | MEDLINE | ID: mdl-11434612

ABSTRACT

BACKGROUND/AIMS: Chronic intoxication of mice with 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) or griseofulvin (GF) results in appearance of Mallory bodies (MBs) and alterations of the keratin cytoskeleton, which are reversible upon drug withdrawal but recur after readministration within 2-3 days. METHODS: DDC- or GF-treated and recovered mice were reintoxicated with the original drugs but also colchicine and lumicolchicine. Cytoskeletal alterations of hepatocytes and MB formation were monitored by immunofluorescence microscopy using keratin, MB-specific antibodies, antibodies to phosphoepitopes and to HSP70. Keratin 8/18 mRNA expression and protein levels were determined by competitive reverse transcription-polymerase chain reaction, in situ-hybridization and western blotting. RESULTS: Duration of pretreatment was important for the efficiency of MB triggering. Rapid increase of keratin 8/18 mRNA and proteins was found in all reintoxicated mice concomitant with MB formation, whereby keratin 8 prevailed over keratin 18. Keratins and a protein with heat shock characteristics (M(M) 120-1 antigen) were the earliest detectable MB components, whereas ubiquitination and phosphorylation followed later. CONCLUSIONS: Overproduction of keratins is a major but not the only step responsible for MB formation. Additional components (e.g. M(M) 120-1 antigen) and excess of keratin 8 over keratin 18 are essential.


Subject(s)
Inclusion Bodies/physiology , Liver/physiology , Animals , Antigens, Surface/pharmacology , Colchicine/pharmacology , Dicarbethoxydihydrocollidine/poisoning , Griseofulvin/poisoning , Immunohistochemistry , Keratin-8 , Keratins/genetics , Keratins/metabolism , Liver/drug effects , Liver/metabolism , Lumicolchicines/pharmacology , Male , Mice , Phosphorylation , RNA, Messenger/metabolism , Time Factors , Ubiquitin/metabolism
6.
Gastroenterology ; 121(1): 170-83, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11438506

ABSTRACT

BACKGROUND AND AIMS: Cholestasis is associated with retention of potentially toxic bile acids and alterations in hepatocellular transporter expression. Conversely, nontoxic ursodeoxycholic acid (UDCA) stimulates bile secretion and counteracts cholestasis. This study aimed to determine the effects of UDCA and cholic acid (CA) on the expression of hepatocellular transporters for bile acids (Ntcp, Bsep), organic anions (Oatp1, Mrp2), organic cations (Mdr1a/b), and phospholipids (Mdr2) in mouse liver. METHODS: Bile flow/composition was analyzed in UDCA- or CA-fed mice. Transporter expression was studied by reverse-transcription polymerase chain reaction, Western blotting, and immunofluorescence microscopy. RESULTS: UDCA had no effect on basolateral Ntcp and down-regulated Oatp1, whereas canalicular Bsep and Mrp2 were up-regulated. CA down-regulated basolateral Ntcp and Oatp1, whereas canalicular Bsep, Mrp2, and Mdr1a/b were up-regulated. Neither UDCA nor CA affected Mdr2 expression. Both UDCA and CA stimulated biliary bile acid and glutathione excretion, although only CA increased phospholipid and cholesterol excretion. CONCLUSIONS: Down-regulation of basolateral and up-regulation of canalicular transporters in response to CA may represent a defense mechanism, in an attempt to prevent hepatocellular accumulation of potentially toxic bile acids. The therapeutic effects of UDCA may be caused in part by stimulation of canalicular transporter expression in the absence of hepatocellular toxicity.


Subject(s)
Bile Ducts/metabolism , Cholic Acid/pharmacology , Liver/drug effects , Ursodeoxycholic Acid/pharmacology , Administration, Oral , Animals , Bile Acids and Salts/metabolism , Biological Transport/drug effects , Cholic Acid/administration & dosage , Down-Regulation/drug effects , Gene Expression Regulation/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects , Ursodeoxycholic Acid/administration & dosage
7.
Hepatology ; 33(3): 633-46, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11230744

ABSTRACT

Reduced hepatobiliary transporter expression could explain impaired hepatic uptake and excretion of bile salts and other biliary constituents resulting in cholestasis and jaundice. Because little is known about alterations of hepatobiliary transport systems in human cholestatic liver diseases, it was the aim of this study to investigate such potential changes. Hepatic mRNA levels in hepatobiliary transport systems for bile salts (NTCP, BSEP), organic anions (OATP2, MRP2, MRP3), organic cations (MDR1), phospholipids (MDR3), and aminophospholipids (FIC1) were determined in 37 human liver biopsies and control livers by competitive reverse-transcription polymerase chain reaction (RT-PCR). Transporter tissue distribution was investigated by immunofluorescence microscopy. In patients with inflammation-induced icteric cholestasis (mainly cholestatic alcoholic hepatitis), mRNA levels of NTCP, OATP2, and BSEP were reduced by 41% (P <.001), 49% (P <.005), and 34% (P <.05) compared with controls, respectively. In addition, NTCP and BSEP immunostaining was reduced. MRP2 mRNA levels remained unchanged, but canalicular immunolabeling for MRP2 was also decreased. mRNA expression of MRP3, MDR1, MDR3, and FIC1 remained unchanged. In contrast to the alterations of transporter expression in inflammation-induced icteric cholestasis, transporter expression did not change in anicteric cholestasis caused by primary biliary cirrhosis (PBC) stages I and II. In conclusion, reduced expression of hepatobiliary transport systems for bile salts and other organic anions may contribute to inflammation-induced cholestasis in humans. Reduction of transporter gene expression can occur at the mRNA level as observed for NTCP, OATP2, and BSEP. However, reduced MRP2 immunostaining in the presence of conserved MRP2 mRNA levels suggests an additional role for posttranscriptional/posttranslational mechanisms.


Subject(s)
Bile Ducts/metabolism , Carrier Proteins/metabolism , Cholestasis/metabolism , Liver/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphatases/genetics , Adult , Anions/metabolism , Bile Acids and Salts/metabolism , Biopsy , Carrier Proteins/genetics , Cholestasis/pathology , Female , Fluorescent Antibody Technique , Humans , Liver/pathology , Male , Middle Aged , RNA, Messenger/metabolism , Reference Values
8.
Pathologe ; 22(6): 388-98, 2001 Nov.
Article in German | MEDLINE | ID: mdl-11766638

ABSTRACT

Alcoholic (ASH) and nonalcoholic (NASH) steatohepatitis show an almost identical morphology. Since the clinical picture is not characteristic, liver biopsy is still the diagnostic gold standard. ASH and NASH are morphologically characterized by a combination of steatosis, hepatocellular injury (ballooning degeneration, apoptosis, necrosis), perivenular and pericellular fibrosis, and inflammation (mostly neutrophils). A definitive differentiation of ASH and NASH is only possible by exclusion of alcohol abuse. Although NASH comprises a syndrome with a multifactorial etiology, adipositas seems to be the most constant associated causal factor. The pathogenesis of both diseases is still unclear. Clinical evidence and experimental studies suggest an important toxic role of reactive oxygen species (oxidative stress). According to our experience, ballooning of hepatocytes is a constant morphologic feature of ASH and NASH and already present in the early stages of disease. Ballooned cells often (but not always) contain Mallory bodies (alcoholic hyalin), which are irregular cytoplasmic inclusions consisting of keratins and nonkeratin components, including ubiquitin. Ballooning is associated with a disturbance and finally almost disappearance of the keratin-intermediate filament cytoskeleton. In our studies on the pathogenesis of ASH and NASH, we concentrated on these cytoskeletal alterations and Mallory body formation. It could be shown that in the early stages overexpression and hyperphosphorylation of keratins take place. Moreover, the 1:1 ratio of keratin type I (keratin 18) and type II (keratin 8) necessary for the assembly of intermediate filaments is disturbed and the equilibrium shifted toward keratin 8. Thus, the pool of soluble keratin 8 increases. The resulting keratin monomers are sensitive to misfolding and either degraded or aggregated as inclusion bodies. If the proteolytic capacity is impaired (e.g., by inhibition of the proteasomal system) in the chronically stressed cell aggregation prevails,finally leading to Mallory body formation. Convincing evidence exists on the basis of clinical and experimental studies that keratins exert a nonskeletal protective function in simple epithelia (e.g., liver cells). Disturbance of the keratin system may thus significantly contribute to cell damage.


Subject(s)
Fatty Liver, Alcoholic/pathology , Fatty Liver/pathology , Biopsy , Fatty Liver/etiology , Humans , Inclusion Bodies/pathology , Keratins/analysis , Liver/pathology
9.
J Cell Biol ; 145(5): 1049-61, 1999 May 31.
Article in English | MEDLINE | ID: mdl-10352021

ABSTRACT

Mice lacking the AP-1 transcription factor c-Jun die around embryonic day E13.0 but little is known about the cell types affected as well as the cause of embryonic lethality. Here we show that a fraction of mutant E13.0 fetal livers exhibits extensive apoptosis of both hematopoietic cells and hepatoblasts, whereas the expression of 15 mRNAs, including those of albumin, keratin 18, hepatocyte nuclear factor 1, beta-globin, and erythropoietin, some of which are putative AP-1 target genes, is not affected. Apoptosis of hematopoietic cells in mutant livers is most likely not due to a cell-autonomous defect, since c-jun-/- fetal liver cells are able to reconstitute all hematopoietic compartments of lethally irradiated recipient mice. A developmental analysis of chimeras showed contribution of c-jun-/- ES cell derivatives to fetal, but not to adult livers, suggesting a role of c-Jun in hepatocyte turnover. This is in agreement with the reduced mitotic and increased apoptotic rates found in primary liver cell cultures derived from c-jun-/- fetuses. Furthermore, a novel function for c-Jun was found in heart development. The heart outflow tract of c-jun-/- fetuses show malformations that resemble the human disease of a truncus arteriosus persistens. Therefore, the lethality of c-jun mutant fetuses is most likely due to pleiotropic defects reflecting the diversity of functions of c-Jun in development, such as a role in neural crest cell function, in the maintenance of hepatic hematopoiesis and in the regulation of apoptosis.


Subject(s)
Heart/embryology , Heart/physiology , Liver/embryology , Liver/physiology , Proto-Oncogene Proteins c-jun/physiology , Transcription Factor AP-1/physiology , Animals , Apoptosis , Embryonic and Fetal Development , Gene Deletion , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/physiology , Liver/pathology , Mice , Mice, Knockout
10.
Am J Pathol ; 154(6): 1701-10, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10362795

ABSTRACT

Intracytoplasmic hyaline bodies (IHBs) resemble inclusions in hepatocellular carcinoma cells, which so far have escaped further characterization. A relationship to Mallory bodies was suggested on the basis of light microscopy and filamentous ultrastructure. A hepatocellular carcinoma containing numerous IHBs was studied. Our studies revealed immunoreactivity of IHBs with the monoclonal antibodies SMI 31 and MPM-2, which recognize hyperphosphorylated epitopes present on paired helical filaments in Alzheimer's disease brains (SMI 31) or on diverse proteins hyperphosphorylated by mitotic kinases in the M-phase of the cell cycle (MPM-2). One- and two-dimensional gel electrophoresis of tumor extracts followed by immunoblotting with SMI 31 and MPM-2 antibodies revealed a major immunoreactive protein with an apparent molecular weight between 62 and 65 kd, which was resolved into several highly acidic (pH 4.5) protein components in two-dimensional gels. This protein was undetectable in non-neoplastic liver tissue. Sequence analysis identified the SMI 31 and MPM-2 immunoreactive material as p62, indicating that p62 is a major constituent of IHBs. p62 is an only recently discovered protein that is a phosphotyrosine-independent ligand of the SH2 domain of p56(lck), a member of the c-src family of cytoplasmic kinases. Moreover, p62 binds ubiquitin and may act as an adapter linking ubiquitinated species to other proteins. These features suggest a role of p62 in signal transduction and possibly also carcinogenesis. IHBs observed in the hepatocellular carcinoma cells presented are the first indications of a role of p62 in disease.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carrier Proteins/metabolism , Hyalin/metabolism , Immediate-Early Proteins/metabolism , Inclusion Bodies/metabolism , Liver Neoplasms/metabolism , Proteins , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Antibodies, Monoclonal , Blotting, Western , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/ultrastructure , Carrier Proteins/immunology , Electrophoresis, Polyacrylamide Gel , Fatal Outcome , Fluorescent Antibody Technique, Indirect , Humans , Hyalin/ultrastructure , Immediate-Early Proteins/immunology , Immunohistochemistry , Inclusion Bodies/ultrastructure , Liver Neoplasms/pathology , Liver Neoplasms/ultrastructure , Male , Microscopy, Electron , Middle Aged , Molecular Sequence Data , Sequence Analysis , Sequestosome-1 Protein
11.
Lab Invest ; 79(2): 103-9, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10068199

ABSTRACT

Many acute and chronic liver diseases are often associated with atypical ductular proliferation (ADP). These ADPs have gained increasing interest since a number of recent observations suggest that ADPs may represent progenies of the putative liver stem cell compartment. In this study, we show that feeding mice with 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) results in persistent proliferation of primitive ductules with poorly defined lumens. Similar to oval cell proliferation in other rodent models as well as in various human liver diseases, DDC-induced ADP originated from the portal tract, spread into the hepatic lobule, and was associated closely with appearance of hepatocytes harboring an antigen (A6), which normally is expressed in biliary epithelium. Furthermore, DDC treatment severely inhibited the regenerative capacity of mice after partial hepatectomy. The development of ADP was selectively blocked in DDC-fed TGF-beta1 transgenic mice producing active TGF-beta1 in the liver and no accumulation of new hepatocytes expressing the A6 antigen was observed. Moreover, the transforming growth factor beta1 (TGF-beta1) transgenic mice did not survive beyond 3 weeks from starting the DDC-containing diet. The results suggest that persistent activation of the hepatic stem cell compartment is essential for liver regeneration in the DDC model and that active TGF-beta1 may negatively control activation of stem cells in the liver. These data further emphasize the relevance of the DDC model as an experimental tool for studying chronic liver diseases.


Subject(s)
Bile Ducts, Intrahepatic/drug effects , Bile Ducts, Intrahepatic/pathology , Liver Diseases, Alcoholic/pathology , Transforming Growth Factor beta/pharmacology , Animals , Cell Division/drug effects , Chemical and Drug Induced Liver Injury , Chronic Disease , Common Bile Duct , Dicarbethoxydihydrocollidine , Disease Models, Animal , Epithelial Cells/pathology , Hepatectomy/methods , Ligation , Liver/drug effects , Liver/pathology , Liver Regeneration/drug effects , Mice , Mice, Transgenic/genetics , Transforming Growth Factor beta/genetics
12.
Verh Dtsch Ges Pathol ; 83: 254-9, 1999.
Article in English | MEDLINE | ID: mdl-10714219

ABSTRACT

Intracytoplasmic hyaline bodies (IHBs) resemble a peculiar type of cytoplasmic inclusions in cells of hepatocellular carcinoma (HCC) which so far have escaped further characterization. In order to determine protein composition of IHBs we investigated tissue of a HCC containing numerous IHBs by immunohistochemistry and Western blot analysis using a large panel of different antibodies. Our studies revealed immunoreactivity of IHBs with the monoclonal antibodies SMI 31 and MPM-2 which recognize hyperphosphorylated epitopes present on paired helical filaments in Alzheimer's disease brains (SMI 31) and on proteins hyperphosphorylated by mitotic kinases (MPM-2), respectively. In two-dimensional Western blots of HCC extracts SMI 31 and MPM-2 antibodies detected a 62 to 65 kD protein with an isoelectric point around 4.5. Microsequencing identified this protein as p62, a recently identified phosphotyrosine-independent ligand of the SH2 domain of tyrosine kinase p56lck. Immunoreactivity of p62 protein spots with antibodies to phosphorylated epitopes (i.e. SMI 31 and MPM-2) suggest that p62 is highly phosphorylated in IHBs. This is the first report on accumulation of p62 as cellular inclusions and its association with human disease.


Subject(s)
Adaptor Proteins, Signal Transducing/analysis , Carcinoma, Hepatocellular/pathology , Inclusion Bodies/pathology , Liver Neoplasms/pathology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Amino Acid Sequence , Antibodies, Monoclonal , Antigens, Neoplasm/analysis , Carcinoma, Hepatocellular/enzymology , Cytoplasm/pathology , Humans , Inclusion Bodies/enzymology , Liver Neoplasms/enzymology , Molecular Sequence Data , Phosphotyrosine/metabolism , Sequestosome-1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...