Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Biotech Histochem ; 92(5): 363-373, 2017.
Article in English | MEDLINE | ID: mdl-28766965

ABSTRACT

Immunohistochemistry (IHC) is used to detect antibody-specific antigens in tissues; the results depend on the ability of the primary antibodies to bind to their antigens. Therefore, results depend on the quality of preservation of the specimen. Many investigators have overcome the deleterious effects of over-fixation on the binding of primary antibodies to specimen antigens using IHC, but if the specimen is under-fixed or fixation is delayed, false negative results could be obtained despite certified laboratory practices. Microtubule-associated protein 2 (MAP2) is an abundant microtubule-associate protein that participates in the outgrowth of neuronal processes and synaptic plasticity; it is localized primarily in cell bodies and dendrites of neurons. MAP2 immunolabeling has been reported to be absent in areas of the entorhinal cortex and hippocampus of Alzheimer's disease brains that were co-localized with the dense-core type of amyloid plaques. It was hypothesized that the lack of MAP2 immunolabeling in these structures was due to the degradation of the MAP2 antigen by the neuronal proteases that were released as the neurons lysed leading to the formation of these plaques. Because MAP2 is sensitive to proteolysis, we hypothesized that changes in MAP2 immunolabeling may be correlated with the degree of fixation of central nervous system (CNS) tissues. We detected normal MAP2 immunolabeling in fixed rat brain tissues, but MAP2 immunolabeling was decreased or lost in unfixed and delayed-fixed rat brain tissues. By contrast, two ubiquitous CNS-specific markers, myelin basic protein and glial fibrillary acidic protein, were unaffected by the degree of fixation in the same tissues. Our observations suggest that preservation of various CNS-specific antigens differs with the degree of fixation and that the lack of MAP2 immunolabeling in the rat brain may indicate inadequate tissue fixation. We recommend applying MAP2 IHC for all CNS tissues as a pre-screen to assess the quality of the tissue preservation and to avoid potentially false negative IHC results.


Subject(s)
Antigens/metabolism , Brain/metabolism , Microtubule-Associated Proteins/metabolism , Animals , Antibodies/metabolism , Brain/immunology , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley , Tissue Fixation/standards
2.
Biotech Histochem ; 85(2): 133-47, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20121465

ABSTRACT

The details of the sequence of pathological events leading to neuron death in Alzheimer's disease (AD) are not known. Even the formation of amyloid plaques, one of the major histopathological hallmarks of AD, is not clearly understood; both the origin of the amyloid and the means of its deposition remain unclear. It is still widely considered, however, that amyloid plaques undergo gradual growth in the interstitial space of the brain via continual extracellular deposition of amyloid beta peptides at "seeding sites," and that these growing plaques encroach progressively on neurons and their axons and dendritic processes, eventually leading to neuronal death. Actually, histopathological evidence to support this mechanism is sparse and of uncertain validity. The fact that the amyloid deposits in AD brains that are collectively referred to as plaques are of multiple types and that each seems to have a different origin often is overlooked. We have shown experimentally that many of the so-called "diffuse amyloid plaques," which lack associated inflammatory cells, are either the result of leaks of amyloid from blood vessels at focal sites of blood-brain barrier breaches or are artifacts resulting from grazing sections through the margins of dense core plaques. In addition, we have provided experimental evidence that neuronal death via necrosis leaves a residue that takes the form of a spheroid "cloud" of amyloid, released by cell lysis, surrounding a dense core that often contains neuronal nuclear material. Support for a neuronal origin for these "dense core amyloid plaques" includes their ability to attract inflammatory cells (microglia and immigrant macrophages) and that they contain nuclear and cytoplasmic components that are somewhat resistant to proteolysis by lysosomes released during neuronal cell lysis. We discuss here the clinical and therapeutic importance of recognizing that amyloid deposition occurs both within neurons (intracellular) and in the interstitial (extracellular) space of the brain. For dense core plaques, we propose that the latter location largely follows from the former. This scenario suggests that blocking intraneuronal amyloid deposition should be a primary therapeutic target. This strategy also would be effective for blocking the gradual compromise of neuronal function resulting from this intraneuronal deposition, and the eventual death and lysis of these amyloid-burdened neurons that leads to amyloid release and the appearance of dense core amyloid plaques in the interstitium of AD brains.


Subject(s)
Alzheimer Disease/pathology , Blood-Brain Barrier/pathology , Brain/pathology , Inflammation/pathology , Neurons/pathology , Plaque, Amyloid/pathology , Aged , Alzheimer Disease/physiopathology , Alzheimer Disease/therapy , Amyloid , Extracellular Space/chemistry , Humans , Immunohistochemistry , Intracellular Space/chemistry , Necrosis , Plaque, Amyloid/classification
3.
Biotech Histochem ; 85(3): 195-204, 2010 Apr 28.
Article in English | MEDLINE | ID: mdl-20109096

ABSTRACT

We previously reported a sensitive, quantitative immunohistochemical assay using formalin fixed, paraffin embedded rat testicular tissues to assess the degree of proliferation-related toxicity. An indexing scheme was devised based on the percentage of PCNA-positive cells positioned as a single layer along the basement membrane at the perimeter of similarly staged seminiferous tubules (PCNA index). We observed significant decreases in the PCNA index in testes of rats treated with an experimental compound that has been shown to produce testicular histopathology. This relatively simple assay provided a more quantitative and sensitive assessment of early testicular toxicity. A separate investigation of the rates of apoptosis in adjacent serial sections of affected rat seminiferous tubules showed that the incidence of apoptosis increased as the rate of proliferation of spermatogonial cells in the tubules decreased. Therefore, we developed a simultaneous PCNA immunohistochemical and TUNEL histochemical assay not only to reduce preparation and analysis time but also to allow determination of the relation between effects of various compounds producing testicular toxicity on the two cellular processes within the same tissue section. We show that an experimental compound known to cause testicular toxicity produced a concurrent reduction of proliferation and increase in apoptosis in seminiferous tubules. In dose-response studies, we show that increased apoptosis was apparent at lower doses that did not show a significant decrease in PCNA, thus indicating the greater sensitivity of the TUNEL indexing assay to detect early evidence of toxicity. Detailed analyses show the presence of TUNEL-positive cells in tubules with normal PCNA labeling, which suggests that an effect on apoptosis occurs prior to significant changes in cell proliferation in the meiotic pathway for this particular testicular toxicant. This single assay employing the simultaneous dual labeling of apoptosis and proliferation has potential utility for detecting early testicular toxicity of experimental compounds in preclinical development and shedding light on potential cellular mechanisms for toxicity, which should help identify compounds with reduced testicular toxicity.


Subject(s)
Proliferating Cell Nuclear Antigen/toxicity , Seminiferous Tubules/drug effects , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , In Situ Nick-End Labeling , Male , Rats , Rats, Sprague-Dawley
4.
Clin Exp Allergy ; 39(8): 1179-86, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19400896

ABSTRACT

BACKGROUND: Secretory leucocyte protease inhibitor (SLPI), which is present in many physiological fluids including saliva, sputum and nasal discharge, is the most effective inhibitor of chymase. Previously, we demonstrated that chymase is able to cleave SLPI and that the cleaved portion, cSLPI, is a biomarker of chymase activity. OBJECTIVE: We investigated the potential of cSLPI as a biomarker of chymase activity in subjects with allergic rhinitis (AR) and asthmatic airway disease. METHODS: Baseline sputum samples were collected from atopic asthmatics and healthy controls (HC). Nasal lavages (NAL) were performed in subjects with AR both at baseline and following a nasal challenge with allergen or placebo. Levels of cSLPI and chymase were determined by Western analysis, and tryptase and alpha-2 macroglobulin were measured by immunoassay. RESULTS: As compared with HC, asthmatics showed a significant increase in baseline cSLPI/total SLPI ratios and an increase in chymase levels. There was a high correlation of cSLPI/SLPI ratios to chymase levels in normal individuals and untreated asthmatics. In the NAL of patients with AR, as compared with placebo, allergen challenge increased inflammatory biomarkers, including cSLPI/SLPI ratios, chymase levels, tryptase levels and alpha2-macroglobulin levels. Correlations were observed between cSLPI/SLPI ratios and chymase levels and cSLPI/SLPI ratios and alpha2-macroglobulin levels; no correlation was seen between cSLPI/SLPI ratios and tryptase levels. CONCLUSION: Our data indicate that cSLPI reflects chymase activity in AR and asthma. Hence, cSLPI may serve as a biomarker for disease activity and for monitoring the efficacy of novel anti-inflammatory treatments in chymase-mediated diseases.


Subject(s)
Chymases/metabolism , Respiratory Hypersensitivity/metabolism , Secretory Leukocyte Peptidase Inhibitor/metabolism , Adolescent , Adult , Aged , Biomarkers/metabolism , Female , Humans , Male , Middle Aged , Nasal Lavage Fluid/chemistry , Nasal Lavage Fluid/immunology , Respiratory Hypersensitivity/enzymology , Respiratory Hypersensitivity/immunology , Retrospective Studies , Rhinitis, Allergic, Seasonal/immunology , Rhinitis, Allergic, Seasonal/metabolism , Secretory Leukocyte Peptidase Inhibitor/immunology , Sputum/enzymology , Sputum/immunology , Sputum/metabolism , Tryptases/metabolism , alpha-Macroglobulins/metabolism
5.
Biotech Histochem ; 83(5): 211-20, 2008 Oct.
Article in English | MEDLINE | ID: mdl-19016366

ABSTRACT

It is well known that toxicants such as cyclophosphamide and ethanol can have deleterious effects on normal spermatogenesis. End points such as testis weight and sperm counts have been used widely to assess gross structural and functional changes in testes resulting from toxicant exposure. Histopathological assessments are more sensitive measures of testicular health, but generally they are neither quantitative nor sensitive enough to detect early toxicity. Recently, immunolabeling cells with proliferating cell nuclear antigen (PCNA) has been used to identify proliferating spermatogonia; however, there have been no systematic attempts to quantify these changes. We have developed a sensitive, reliable and quantitative assay using immunohistochemistry on formalin fixed, paraffin embedded rat testes to assess the degree of proliferation-related toxicity. An indexing scheme was derived based on the determination of radially positioned PCNA-positive cells within similarly staged seminiferous tubules presenting a single layer of PCNA-positive cells along the basement membrane of the basal tubular compartment. An average of 60 tubules in the testes were counted per animal. Our results show significant decreases in the PCNA index in rats treated with an experimental compound that has been shown to produce testicular histopathology. The analysis provides a quick, reliable, sensitive, and quantitative means for assessing early testicular toxicity. The assay has potential utility as an in vivo biomarker for detecting early testicular toxicity of experimental compounds in preclinical development as well as for refining follow-up compounds with reduced testicular toxicity.


Subject(s)
Biomarkers/analysis , Drug Evaluation, Preclinical , Proliferating Cell Nuclear Antigen/analysis , Testis/cytology , Animals , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley , Seminiferous Tubules/chemistry , Sensitivity and Specificity , Spermatogonia/chemistry , Spermatogonia/cytology
6.
Ann Oncol ; 18 Suppl 6: vi141-8, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17591809

ABSTRACT

Historically, treatment options for metastatic renal cell carcinoma (RCC) have been limited because of inherent tumor resistance to chemotherapy and radiotherapy. The only approved drug for RCC in the past 30 years has been high-dose interleukin-2. Its benefit is observed in a small percentage (20%-25%) of highly selected good performance status RCC patients. The treatment of advanced RCC has recently undergone a major change with the development of potent angiogenesis inhibitors and targeted agents. In fact, advanced RCC is a highly vascular tumor associated with expression of vascular endothelial growth factor (VEGF); thereafter, antiangiogenic strategies have become an attractive approach. Several multitargeted tyrosine kinase inhibitors (sorafenib and sunitinib) have already been approved for the treatment of advanced RCC; bevacizumab, a monoclonal antibody anti-VEGF, has shown promising clinical activity. Temsirolimus, a derivative of rapamycin (CCI-779), has also shown a survival advantage over interferon in advanced, poor-prognosis RCC patients. The aim of this review is to describe these agents in terms of mechanisms of action, efficacy, and toxicity profile and also to analyze future development strategies.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/secondary , Carcinoma, Renal Cell/surgery , Combined Modality Therapy , Drug Delivery Systems/adverse effects , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Kidney Neoplasms/surgery , Nephrectomy
7.
Int J Biol Markers ; 22(1 Suppl 4): S88-93, 2007.
Article in English | MEDLINE | ID: mdl-17520586

ABSTRACT

We report here on the state of our knowledge of the target--namely, the epidermal growth factor (EGF) and its receptor--and the challenges related to the methods of determination of the epidermal growth factor receptor (EGFR) and associated molecular pathways. A critical review of the anti-EGFR therapeutic strategies is also outlined. The chimeric anti- EGFR monoclonal antibody cetuximab has been approved for EGFR-expressing colorectal tumors in patients who progress after irinotecan-based chemotherapy in combination with irinotecan and in squamous cell head and neck carcinomas for patients with locally advanced disease in combination with radiation therapy or after failure of platinum-based chemotherapy in recurrent or metastatic disease (FDA). Cetuximab has the potential to provide an improvement of clinical outcome also in other indications and tumor types, particularly when used as first-line therapy combined with standard chemotherapy for metastatic disease or in the adjuvant setting. Possible strategies to improve the effectiveness of anti-EGFR agents are suggested and include (i) the use of predictive tools capable of making a more rational selection of patients; (ii) the development of standardized predictive biomarkers as surrogates for early monitoring of drug efficacy; and (iii) adequate study design, statistical analysis and proper end points of efficacy to be applied in future prospective trials.


Subject(s)
Antineoplastic Agents/therapeutic use , ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/drug therapy , Cetuximab , Colorectal Neoplasms/drug therapy , ErbB Receptors/analysis , ErbB Receptors/genetics , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Lung Neoplasms/drug therapy , Predictive Value of Tests
8.
Int J Biol Markers ; 22(1): 24-33, 2007.
Article in English | MEDLINE | ID: mdl-17393358

ABSTRACT

The present article overviews the role of bisphosphonates for the treatment and prevention of bone metastases and their antiangiogenic effects and antitumoral activity. The skeleton is a frequent and clinically relevant site of metastasis in cancer patients. The major events related to bone metastases include bone pain, bone loss, hypercalcemia, spinal cord compression, and fractures. On the basis of their radiographic features, bone metastases are classified as osteoblastic, osteoclastic, or mixed. The primary goals of treatment of bone metastases are reduction of the risk of pathological fractures and other skeletal-related events, and pain control. Bisphosphonates are used to prevent pathological fractures by inhibition of osteoclasts. Recent studies suggest that bisphosphonates have some direct antitumoral activity, mainly mediated through the blockade of angiogenic pathways. Further clinical studies are needed to determine the optimal treatment duration, timing and schedule of bisphosphonates, assess their role as adjuvant therapy for the prevention of bone metastases, and establish their antiangiogenic activity in association with standard cytotoxic and hormonal drugs for treatment of patients with advanced disease.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Diphosphonates/therapeutic use , Alkaline Phosphatase/analysis , Biomarkers, Tumor/analysis , Bone Resorption/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Clinical Trials as Topic , Clodronic Acid/therapeutic use , Female , Humans , Ibandronic Acid , Imidazoles/therapeutic use , Male , Pamidronate , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Zoledronic Acid
9.
Int J Biol Markers ; 22(4): 88-93, 2007.
Article in English | MEDLINE | ID: mdl-28207119

ABSTRACT

We report here on the state of our knowledge of the target namely, the epidermal growth factor (EGF) and its receptor and the challenges related to the methods of determination of the epidermal growth factor receptor (EGFR) and associated molecular pathways. A critical review of the anti-EGFR therapeutic strategies is also outlined. The chimeric anti- EGFR monoclonal antibody cetuximab has been approved for EGFR-expressing colorectal tumors in patients who progress after irinotecan-based chemotherapy in combination with irinotecan and in squamous cell head and neck carcinomas for patients with locally advanced disease in combination with radiation therapy or after failure of platinum-based chemotherapy in recurrent or metastatic disease (FDA). Cetuximab has the potential to provide an improvement of clinical outcome also in other indications and tumor types, particularly when used as first-line therapy combined with standard chemotherapy for metastatic disease or in the adjuvant setting. Possible strategies to improve the effectiveness of anti-EGFR agents are suggested and include (i) the use of predictive tools capable of making a more rational selection of patients; (ii) the development of standardized predictive biomarkers as surrogates for early monitoring of drug efficacy; and (iii) adequate study design, statistical analysis and proper end points of efficacy to be applied in future prospective trials.

10.
Int J Biol Markers ; 21(2): 111-22, 2006.
Article in English | MEDLINE | ID: mdl-16847814

ABSTRACT

For the past 15 years tamoxifen has been the standard adjuvant hormone therapy for women with early-stage breast cancer and estrogen receptor (ER)-positive tumors, irrespective of nodal status and other clinicopathological parameters. Recent studies provided evidence that the optimal duration of tamoxifen treatment is 5 years. Based on the positive clinical results obtained with the administration of aromatase inhibitors (AIs) in the metastatic setting, several controlled clinical trials have evaluated the efficacy and side effects of AIs versus standard tamoxifen also as adjuvant therapy in postmenopausal breast cancer patients. The results of the above studies, suggest a therapeutic advantage of AIs over tamoxifen with regard to relapse-free survival and the risk of metachronous contralateral breast cancer. We review the rationale and the available clinical data on initial or sequential hormone treatment with AIs and we propose a novel scenario for possible therapeutic strategies based on the clinicopathological characteristics of the patients and on the biology of each single tumor.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/metabolism , Chemotherapy, Adjuvant , Hormones/therapeutic use , Aromatase Inhibitors/pharmacology , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Growth Substances/metabolism , Humans , Models, Biological , Neoplasm Metastasis , Postmenopause , Receptors, Estrogen/metabolism , Risk , Tamoxifen/pharmacology , Treatment Outcome
11.
Neurogastroenterol Motil ; 16(6): 811-8, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15601431

ABSTRACT

Neurogenic mechanisms have been implicated in the induction of inflammatory bowel disease (IBD). Vanilloid receptor type 1 (TRPV1) has been visualized on nerve terminals of intrinsic and extrinsic afferent neurones innervating the gastrointestinal tract and local administration of a TRPV1 antagonist, capsazepine, reduces the severity of dextran sulphate sodium (DSS)-induced colitis in rats (Gut 2003; 52: 713-9(1)). Our aim was to test whether systemically or orally administered TRPV1 antagonists attenuate experimental colitis induced by 5% DSS in Balb/c mice. Intraperitoneal capsazepine (2.5 mg kg(-1), bid), significantly reduced the overall macroscopic damage severity compared with vehicle-treated animals (80% inhibition, P < 0.05); however, there was no effect on myeloperoxidase (MPO) levels. An experimental TRPV1 antagonist given orally was tested against DSS-induced colitis, and shown to reverse the macroscopic damage score at doses of 0.5 and 5.0 mg kg(-1). Epithelial damage assessed microscopically was significantly reduced. MPO levels were attenuated by approximately 50%, and diarrhoea scores were reduced by as much as 70%. These results suggest that pharmacological modulation of TRPV1 attenuates indices of experimental colitis in mice, and that development of orally active TRPV1 antagonists might have therapeutic potential for the treatment of IBD.


Subject(s)
Capsaicin/analogs & derivatives , Colitis/prevention & control , Ion Channels/antagonists & inhibitors , Animals , Anticoagulants/pharmacology , Capsaicin/pharmacology , Colitis/chemically induced , Colitis/pathology , Dextran Sulfate/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/prevention & control , Ion Channels/drug effects , Mice , Mice, Inbred BALB C , Peroxidase/drug effects , Peroxidase/metabolism , TRPV Cation Channels
12.
Biotech Histochem ; 79(2): 55-64, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15513707

ABSTRACT

Reliable immunohistochemical detection of collagen in formalin fixed, paraffin embedded tissues requires protease digestion. While these pan-proteases (pepsin, trypsin, protease K, etc.) enhance collagen detection, they also digest many other tissue proteins and produce poor cellular morphology and unrecognizable cellular structures. Balancing the conditions (protease type, concentration, incubation time and temperature) to digest some, but not all, proteins in a tissue section while optimizing collagen detection requires one to compromise improved collagen immunolabeling with adequate cellular morphology. Furthermore, optimal conditions for digesting tissue proteins to enhance collagen detection vary among tissue types and their fixation. Although brain is not typically subject to these deleterious consequences, structures such as epithelium, spermatids, stroma etc. and other tissues with complicated histology are profoundly affected. To resolve this technical dilemma, we discovered a novel use for collagenase to enhance collagen immunodetection without affecting the noncollagen proteins, thereby preserving tissue morphology. Collagenase, which is typically used in vitro for disassociation of cells, has never been used reliably on formalin fixed, paraffin embedded tissue sections. This new use of collagenase for immunohistochemistry promotes increased collagen immunolabeling, is easy to use, is versatile, and allows preservation of tissue structure that provides maximal and accurate histological information.


Subject(s)
Cells, Cultured/cytology , Cells, Cultured/metabolism , Collagen/metabolism , Collagen/ultrastructure , Collagenases/metabolism , Paraffin Embedding/methods , Staining and Labeling/methods , Cell Size , Humans , Immunohistochemistry
13.
Biotech Histochem ; 77(2): 95-103, 2002 Mar.
Article in English | MEDLINE | ID: mdl-12083391

ABSTRACT

Alzheimer's disease (AD) neuropathology is characterized by the presence of diffuse and dense-core (neuritic) amyloid plaques in specific areas of the brain. The origin of these plaques and the relationship between them is poorly understood. Current methods to identify clearly these types of plaques in the AD brains are largely dependent upon morphological characteristics. Dense-core amyloid plaques in the entorhinal cortex and hippocampus of AD brains might arise from the lysis of neurons overburdened by excessive intracellular deposition of amyloid beta1-42 (Abeta42) peptide. The local release of active lysosomal enzymes, which persist within these plaques, might degrade most of the released intracellular proteins, leaving behind only those that are resistant to proteolytic digestion, such as ubiquitin, tau, neurofilament proteins and amyloid. To test the possibility that proteins that are sensitive to proteolysis may be degraded selectively in plaques, we used immunohistochemistry to examine the distribution of microtubule-associated protein-2 (MAP-2), a protein localized primarily in neuronal dendrites and known to be sensitive to proteolysis. Uniform MAP-2 immunolabeling was detected throughout the somatodendritic compartment of neurons in age-matched control cortical brain tissues as well as throughout areas of Abeta42-positive diffuse plaques in AD brains. In contrast, analysis of serial sections revealed that MAP-2 was absent from Abeta42-positive dense-core plaques in AD brains. Our results indicate that this differential MAP-2 immunolabeling pattern among plaques may be employed as a reliable and sensitive method to distinguish dense-core plaques from diffuse plaques within AD brain tissue. Furthermore, this biochemical distinction indicates that dense-core and diffuse plaques are formed by different mechanisms.


Subject(s)
Entorhinal Cortex/pathology , Hippocampus/pathology , Microtubule-Associated Proteins/analysis , Plaque, Amyloid/classification , Plaque, Amyloid/pathology , Aged , Alzheimer Disease/pathology , Biomarkers , Humans , Immunohistochemistry/methods
14.
Neuroscience ; 110(2): 199-211, 2002.
Article in English | MEDLINE | ID: mdl-11958863

ABSTRACT

Amyloid beta(1-42), a major component of amyloid plaques, binds with exceptionally high affinity to the alpha 7 nicotinic acetylcholine receptor and accumulates intracellularly in neurons of Alzheimer's disease brains. In this study, we investigated the possibility that this binding plays a key role in facilitating intraneuronal accumulation of amyloid beta(1-42). Consecutive section immunohistochemistry and digital imaging were used to reveal the spatial relationship between amyloid beta(1-42) and the alpha 7 receptor in affected neurons of Alzheimer's disease brains. Results showed that neurons containing substantial intracellular accumulations of amyloid beta(1-42) invariably express relatively high levels of the alpha 7 receptor. Furthermore, this receptor is highly co-localized with amyloid beta(1-42) within neurons of Alzheimer's disease brains. To experimentally test the possibility that the binding interaction between exogenous amyloid beta(1-42) and the alpha 7 receptor facilitates internalization and intracellular accumulation of amyloid beta(1-42) in Alzheimer's disease brains, we studied the fate of exogenous amyloid beta(1-42) and its interaction with the alpha 7 receptor in vitro using cultured, transfected neuroblastoma cells that express elevated levels of this receptor. Transfected cells exhibited rapid binding, internalization and accumulation of exogenous amyloid beta(1-42), but not amyloid beta(1-40). Furthermore, the rate and extent of amyloid beta(1-42) internalization was related directly to the alpha 7 receptor protein level, since (1) the rate of amyloid beta(1-42) accumulation was much lower in untransfected cells that express much lower levels of this receptor and (2) internalization was effectively blocked by alpha-bungarotoxin, an alpha 7 receptor antagonist. As in neurons of Alzheimer's disease brains, the alpha 7 receptor in transfected cells was precisely co-localized with amyloid beta(1-42) in prominent intracellular aggregates. Internalization of amyloid beta(1-42) in transfected cells was blocked by phenylarsine oxide, an inhibitor of endocytosis. We suggest that the intraneuronal accumulation of amyloid beta(1-42) in Alzheimer's disease brains occurs predominantly in neurons that express the alpha 7 receptor. In addition, internalization of amyloid beta(1-42) may be facilitated by the high-affinity binding of amyloid beta(1-42) to the alpha 7 receptor on neuronal cell surfaces, followed by endocytosis of the resulting complex. This provides a plausible explanation for the selective vulnerability of neurons expressing the alpha 7 receptor in Alzheimer's disease brains and for the fact that amyloid beta(1-42) is the dominant amyloid beta peptide species in intracellular accumulations and amyloid plaques.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Entorhinal Cortex/metabolism , Hippocampus/metabolism , Neurons/metabolism , Peptide Fragments/metabolism , Plaque, Amyloid/metabolism , Receptors, Nicotinic/metabolism , Aged , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/pharmacology , Binding Sites/physiology , Cytoplasm/metabolism , Cytoplasm/pathology , Endocytosis , Entorhinal Cortex/pathology , Entorhinal Cortex/physiopathology , Fluorescent Antibody Technique , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Neurons/drug effects , Neurons/pathology , Peptide Fragments/pharmacology , Plaque, Amyloid/pathology , Receptors, Nicotinic/genetics , Tumor Cells, Cultured , alpha7 Nicotinic Acetylcholine Receptor
15.
Biochemistry (Mosc) ; 67(1): 56-64, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11841340

ABSTRACT

The serine protease thrombin is well recognized as being pivotal to the maintenance of hemostasis under both normal and pathological conditions. Its cellular actions are mediated through a unique family of protease-activated receptors (PARs). These receptors represent a novel family of G protein-coupled receptors that undergo proteolytic cleavage of their amino terminus and subsequent autoactivation by a tethered peptide ligand. This paper reviews the consequences of PAR activation in thrombosis, vascular injury, inflammation, tissue injury, and within the tumor microenvironment.


Subject(s)
Endopeptidases/metabolism , Receptors, Thrombin/metabolism , Thrombin/metabolism , Animals , Humans , Models, Biological , Neoplasms/metabolism , Receptor, PAR-1 , Thrombosis
16.
J Cell Physiol ; 189(3): 341-55, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11748592

ABSTRACT

Many G-protein coupled receptors (GPCRs) undergo ligand-dependent internalization upon activation. The parathyroid hormone (PTH) receptor undergoes endocytosis following prolonged exposure to ligand although the ultimate fate of the receptor following internalization is largely unknown. To investigate compartmentalization of the PTH receptor, we have established a stable cell line expressing a PTH receptor-green fluorescent protein (PTHR-GFP) conjugate and an algorithm to quantify PTH receptor internalization. HEK 293 cells expressing the PTHR-GFP were compared with cells expressing the wild-type PTH receptor in whole-cell binding and functional assays. 125I-PTH binding studies revealed similar Bmax and kD values in cells expressing either the PTHR-GFP or the wild-type PTH receptor. PTH-induced cAMP accumulation was similar in both cell lines suggesting that addition of the GFP to the cytoplasmic tail of the PTH receptor does not alter the ligand binding or G-protein coupling properties of the receptor. Using confocal fluorescence microscopy, we demonstrated that PTH treatment of cells expressing the PTHR-GFP conjugate produced a time-dependent redistribution of the receptor to the endosomal compartment which was blocked by pretreatment with PTH antagonist peptides. Treatment with hypertonic sucrose prevented PTH-induced receptor internalization, suggesting that the PTH receptor internalizes via a clathrin-dependent mechanism. Moreover, co-localization with internalized transferrin showed that PTHR-GFP trafficking utilized the endocytic recycling compartment. Experiments using cycloheximide to inhibit protein synthesis demonstrated that recycling of the PTHR-GFP back to the plasma membrane was complete within 1-2 h of ligand removal and was partially blocked by pretreatment with cytochalasin D, but not nocodazole. We also demonstrated that the PTH receptor, upon recycling to the plasma membrane, is capable of undergoing a second round of internalization, a finding consistent with a role for receptor recycling in functional resensitization.


Subject(s)
Luminescent Proteins/metabolism , Receptors, Parathyroid Hormone/metabolism , Algorithms , Cell Line , Cyclic AMP/biosynthesis , Cytochalasin D/pharmacology , Cytoskeleton/drug effects , Endocytosis , Endosomes/chemistry , Green Fluorescent Proteins , Humans , Indicators and Reagents/metabolism , Luminescent Proteins/genetics , Microscopy, Confocal , Microscopy, Fluorescence , Parathyroid Hormone/metabolism , Parathyroid Hormone/pharmacology , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Transport , Receptors, Parathyroid Hormone/agonists , Receptors, Parathyroid Hormone/genetics , Recombinant Fusion Proteins/metabolism , Transferrin/analysis
17.
Br J Pharmacol ; 133(7): 975-87, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11487506

ABSTRACT

Thrombin, generated in the circulation during injury, cleaves proteinase-activated receptor 1 (PAR1) to stimulate plasma extravasation and granulocyte infiltration. However, the mechanism of thrombin-induced inflammation in intact tissues is unknown. We hypothesized that thrombin cleaves PAR1 on sensory nerves to release substance P (SP), which interacts with the neurokinin 1 receptor (NK1R) on endothelial cells to cause plasma extravasation. PAR1 was detected in small diameter neurons known to contain SP in rat dorsal root ganglia by immunohistochemistry and in situ hybridization. Thrombin and the PAR1 agonist TFLLR-NH(2) (TF-NH(2)) increased [Ca(2+)](i) >50% of cultured neurons (EC(50)s 24 mu ml(-1) and 1.9 microM, respectively), assessed using Fura-2 AM. The PAR1 agonist completely desensitized responses to thrombin, indicating that thrombin stimulates neurons through PAR1. Injection of TF-NH(2) into the rat paw stimulated a marked and sustained oedema. An NK1R antagonist and ablation of sensory nerves with capsaicin inhibited oedema by 44% at 1 h and completely by 5 h. In wild-type but not PAR1(-/-) mice, TF-NH(2) stimulated Evans blue extravasation in the bladder, oesophagus, stomach, intestine and pancreas by 2 - 8 fold. Extravasation in the bladder, oesophagus and stomach was abolished by an NK1R antagonist. Thus, thrombin cleaves PAR1 on primary spinal afferent neurons to release SP, which activates the NK1R on endothelial cells to stimulate gap formation, extravasation of plasma proteins, and oedema. In intact tissues, neurogenic mechanisms are predominantly responsible for PAR1-induced oedema.


Subject(s)
Capillary Permeability/physiology , Neurons/physiology , Receptors, Thrombin/metabolism , Animals , Calcium/metabolism , Capillary Permeability/drug effects , Edema/chemically induced , Edema/metabolism , Esophagus/blood supply , Esophagus/drug effects , Esophagus/innervation , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Jejunum/blood supply , Jejunum/drug effects , Jejunum/innervation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurokinin-1 Receptor Antagonists , Neurons/drug effects , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Oligopeptides/pharmacology , Pancreas/blood supply , Pancreas/drug effects , Pancreas/innervation , Piperidines/pharmacology , Quinuclidines/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptor, PAR-1 , Receptors, Thrombin/agonists , Receptors, Thrombin/chemistry , Signal Transduction/drug effects , Stomach/blood supply , Stomach/drug effects , Stomach/innervation , Thrombin/pharmacology , Urinary Bladder/blood supply , Urinary Bladder/drug effects , Urinary Bladder/innervation
18.
Infect Immun ; 69(8): 5121-30, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11447194

ABSTRACT

Periodontitis is a chronic inflammatory disease affecting oral tissues. Oral epithelial cells represent the primary barrier against bacteria causing the disease. We examined the responses of such cells to an arginine-specific cysteine proteinase (RgpB) produced by a causative agent of periodontal disease, Porphyromonas gingivalis. This protease caused an intracellular calcium transient in an oral epithelial cell line (KB), which was dependent on its enzymatic activity. Since protease-activated receptors (PARs) might mediate such signaling, reverse transcription-PCR was used to characterize the range of these receptors expressed in the KB cells. The cells were found to express PAR-1, PAR-2, and PAR-3, but not PAR-4. In immunohistochemical studies, human gingival epithelial cells were found to express PAR-1, PAR-2, and PAR-3 on their surface, but not PAR-4, indicating that the cell line was an effective model for the in vivo situation. PAR-1 and PAR-2 expression was confirmed in intracellular calcium mobilization assays by treatment of the cells with the relevant receptor agonist peptides. Desensitization experiments strongly indicated that signaling of the effects of RgpB was occurring through PAR-1 and PAR-2. Studies with cells individually transfected with each of these two receptors confirmed that they were both activated by RgpB. Finally, it was shown that, in the oral epithelial cell line, PAR activation by the bacterial protease-stimulated secretion of interleukin-6. This induction of a powerful proinflammatory cytokine suggests a mechanism whereby cysteine proteases from P. gingivalis might mediate inflammatory events associated with periodontal disease on first contact with a primary barrier of cells.


Subject(s)
Cysteine Endopeptidases/metabolism , Endopeptidases/metabolism , Hemagglutinins/metabolism , Interleukin-6/metabolism , Porphyromonas gingivalis/enzymology , Receptors, Thrombin/metabolism , Adhesins, Bacterial , Animals , CHO Cells , Calcium/metabolism , Cell Line , Cricetinae , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression , Gingipain Cysteine Endopeptidases , Gingiva/cytology , Gingiva/metabolism , Humans , Mouth Mucosa/cytology , Mouth Mucosa/metabolism , Receptor, PAR-1 , Receptor, PAR-2 , Receptors, Thrombin/genetics
19.
Am J Pathol ; 158(6): 2031-41, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11395381

ABSTRACT

The serine proteases thrombin and trypsin are among many factors that malignant cells secrete into the extracellular space to mediate metastatic processes such as cellular invasion, extracellular matrix degradation, angiogenesis, and tissue remodeling. The degree of protease secretion from malignant cells has been correlated to their metastatic potential. Protease activated receptors (PAR)-1 and -2, which are activated by thrombin and trypsin respectively, have not been extensively characterized in human tumors in situ. We investigated the presence of PAR-1 and PAR-2 in human normal, benign and malignant tissues using immunohistochemistry and in situ hybridization. Our results demonstrate PAR-1 and PAR-2 expression in the tumor cells, mast cells, macrophages, endothelial cells, and vascular smooth muscle cells of the metastatic tumor microenvironment. Most notably, an up-regulation of PAR-1 and PAR-2 observed in proliferating, smooth muscle actin (SMA)-positive stromal fibroblasts surrounding the carcinoma cells was not observed in normal or benign conditions. Furthermore, in vitro studies using proliferating, SMA-positive, human dermal fibroblasts, and scrape-wounded human dermal fibroblasts demonstrated the presence of PAR-1 and PAR-2 not detected in quiescent, SMA-negative cultures. PAR-1 and PAR-2 in the cells forming the tumor microenvironment suggest that these receptors mediate the signaling of secreted thrombin and trypsin in the processes of cellular metastasis.


Subject(s)
Fibroblasts/metabolism , Neoplasms/metabolism , Receptors, Thrombin/biosynthesis , Stromal Cells/metabolism , Actins/analysis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Cell Division , Cells, Cultured , Female , Fibroblasts/cytology , Humans , Immunohistochemistry , In Situ Hybridization , Neoplasms/genetics , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Receptor, PAR-1 , Receptor, PAR-2 , Receptors, Thrombin/genetics
20.
Neurosci Lett ; 306(3): 137-40, 2001 Jun 29.
Article in English | MEDLINE | ID: mdl-11406314

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder associated with the appearance of cytoplasmic Lewy bodies (LBs) in dopaminergic neurons of the substantia nigra and the progressive loss of these neurons. Cytoskeleton alterations and associated impairments of neuronal transport may contribute to neuronal death. Microtubule-associated protein-2 (MAP-2), a cytoskeleton protein is localized primarily in neuronal dendrites and is known to stabilize microtubule assembly and mediate their interactions with other neuronal cell components. To determine if alterations in MAP-2 morphology are present in PD neurons, we used single and double immunohistochemical and immunofluorescent techniques to characterize MAP-2 in PD neuronal tissues. We report abnormal MAP-2 immunolabeling in some neurons of the substantia nigra of PD brain tissues, which were not observed in the normal, age-matched, control brain tissues. Furthermore, MAP-2 was co-localized with alpha-synuclein and ubiquitin in cytoplasmic LBs of neurons. Surprisingly, MAP-2 was also found to form fibrous aggregates and crystal-like structures within neuronal nuclei. These PD-associated alterations in MAP-2 morphology and distribution suggest that impaired neuronal transport may contribute to the progression of neuronal loss in the brains of PD patients.


Subject(s)
Microtubule-Associated Proteins/analysis , Parkinson Disease/pathology , Substantia Nigra/chemistry , Substantia Nigra/pathology , Aged , Antibodies , Biomarkers , Cell Nucleus/chemistry , Cell Nucleus/pathology , Fluorescent Antibody Technique , Humans , Lewy Bodies/chemistry , Lewy Bodies/pathology , Microtubule-Associated Proteins/immunology , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/immunology , Neurons/chemistry , Neurons/pathology , Synucleins , Ubiquitins/analysis , alpha-Synuclein
SELECTION OF CITATIONS
SEARCH DETAIL
...