Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Article in English | MEDLINE | ID: mdl-38198703

ABSTRACT

Objective: To assess the quality of sleep among patients with schizophrenia.Methods: A cross-sectional descriptive study was conducted with 100 outpatients with schizophrenia recruited from a tertiary care center in Northern India from July 2022 to December 2022. Eligible participants were required to complete a demographic form, the Pittsburgh Sleep Quality Index (PSQI), and the Insomnia Severity Index (ISI). The severity of psychosis was assessed with the Positive and Negative Syndrome Scale (PANSS).Results: The mean age of the participants was 38.87 years (SD = 10.564). The prevalence of poor sleep quality (PSQI ≥ 5) among the patients with schizophrenia was 78%, and the mean PSQI score was 11.2 (SD = 5.31). Most of the participants in whom the prevalence of poor sleep quality was marginally higher were female (51%). Of the 7 PSQI components, daytime dysfunction was more affected (49%) compared to the other 6 components. There was a positive correlation between PSQI and ISI scores (r = 0.805, P < .001). PSQI (U = 380, P < .001) and ISI scores (U = 517, P < .001) were significantly lower in schizophrenia patients taking benzodiazepines. The PANSS scores were not significantly correlated with PSQI or ISI scores.Conclusions: Most patients with schizophrenia suffered significantly from poor sleep quality. The results showed the deleterious impact of poor sleep on their daytime functioning, suggesting the need for comprehensive management of sleep problems in such patients.Prim Care Companion CNS Disord 2023;25(6):23m03564. Author affiliations are listed at the end of this article.


Subject(s)
Schizophrenia , Sulfonamides , Humans , Female , Adult , Male , Schizophrenia/complications , Schizophrenia/epidemiology , Sleep Quality , Tertiary Care Centers , Cross-Sectional Studies , Outpatients , India/epidemiology
2.
J Family Med Prim Care ; 12(12): 3356-3361, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38361839

ABSTRACT

Objective: To find sexual dysfunction in acute-phase bipolar depression patients and subsequently characterize the gender-wise differences in sexual functioning. Materials and Methods: A cross-sectional, descriptive, observational, purposeful, and hospital-based study was done with 45 patients (age range: 18-59 years) with moderate to severe acute phase bipolar depression (HAM-D scores >18). The domain-wise (Pleasure, Desire/Frequency Desire/Interest, Arousal/Excitement, and Orgasm/Completion) sexual functioning was assessed by the Change in Sexual Functioning Questionnaire (CSFQ-14) (≤41 for females, ≤47 for males as a cut-off for dysfunction). This study is registered in the CTRI (Clinical Trials Registry India, Number: CTRI-2021-07-035182). Results: The prevalence of sexual dysfunction was 91% of bipolar disorder patients with more male participants (53.3%) compared to females (46.7%). The mean HAM-D score for the study sample was 27.93 ± 8.035. The female gender had more dysfunctional scores in desire/frequency (t = 2.229, P = 0.031), desire/interest (t = 2.448, P = 0.019), orgasm/completion (t = 2.974, P = 0.005), and overall total CSFQ (t = 2.946, P = 0.005). The odds of sexual dysfunction were significant given a one-unit increase in suicidal ideation in the index episode (adjusted OR = 1.222, 95% CI: 1.004-1.488, P = .049). Conclusion: Acute-phase bipolar patients have very high sexual dysfunction rates. Females have both global and specific sexual response cycle deficits in comparison to acute phase bipolar depressed males. Future trials shall amuse neurobiology grounded, more individualistic sexual rehabilitation-based interventional paradigms, and longitudinal research models in acute phase bipolar depression.

3.
Mol Carcinog ; 58(1): 102-112, 2019 01.
Article in English | MEDLINE | ID: mdl-30302860

ABSTRACT

Despite effective surgical methods for non-melanoma skin cancer (NMSC), patients suffer from tissue damage, scarring, or even disfigurement; thus, there is a need for chemopreventive approaches. Because of the complex interplay between glucocorticoids (GCs), inflammation, and cancer, we sought to determine the role of 11ß-hydroxysteroid dehydrogenase 1 and 2 (11ßHSD1 and 2) in regulating GCs during skin cancer development and progression. 11ßHSDs modulate the activation of GCs in a tissue-specific manner and have been reported to play a role in development and progression of other types of cancer, but their role has not yet been reported in NMSC. Here, we found a significant upregulation of 11ßHSD2 protein in skin cancer cells when compared to normal skin cells, suggesting a role for this enzyme in the multifactorial process of skin cancer development. In addition, inhibition of 11ßHSD2 with siRNA resulted in significant reduction in colony formation in vitro. Finally, our in vivo study elucidated that inhibition of 11ßHSD2 with pharmacological inhibitor, Glycyrrhetinic acid (GA) could significantly diminish tumorigenesis in a well-studied in vivo mouse model of NMSC. Overall, these studies highlight for the first time a potential novel role for 11ßHSD2 in NMSC development and may allow for new GC treatment approaches capable of avoiding deactivation by the enzyme. If 11ßHSD2 can be inhibited as we have done here, or circumvented using modified GCs, this may lead to more efficacious outcomes for NMSC patients by preventing deactivation of the GC and minimizing resistance.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Glucocorticoids/pharmacology , Glycyrrhetinic Acid/pharmacology , Skin Neoplasms/prevention & control , Animals , Apoptosis , Cell Proliferation , Female , Humans , Mice , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Tumor Cells, Cultured
4.
Mol Carcinog ; 57(12): 1698-1706, 2018 12.
Article in English | MEDLINE | ID: mdl-30129681

ABSTRACT

The phytonutrient ursolic acid (UA), present in apples, rosemary, and other plant sources, has anti-cancer properties in a number of systems, including skin cancers. However, few reports have examined upstream mechanisms by which UA may prevent or treat cancer. Recent reports have indicated UA induces death of cancer cell lines via AMP-activated protein kinase (AMPK), an energy-sensing kinase which possesses both pro-metabolic and anti-cancer effects. Other studies have shown UA activates peroxisome proliferator activated receptor α (PPARα) and the glucocorticoid receptor (GR). Here, we found the cytotoxic effect of UA in skin carcinoma cells required AMPK activation. In addition, two inhibitors of PPARα partially reversed the cytotoxic effects of UA, suggesting its effects are at least partially mediated through this receptor. Finally, inhibition of the GR did not reverse the effects of UA nor did this compound bind the GR under the conditions of experiments performed. Overall, studies elucidating the anti-cancer effects of UA may allow for the development of more potent analogues utilizing similar mechanisms. These studies may also reveal the mediators of any possible side effects or resistance mechanisms to UA therapy.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , PPAR alpha/metabolism , Skin Neoplasms/metabolism , Triterpenes/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Signal Transduction/drug effects , Skin Neoplasms/drug therapy , Ursolic Acid
5.
Melanoma Res ; 25(2): 103-12, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25647735

ABSTRACT

Malignant melanoma is associated with a 5-year survival rate of less than 20% once metastasized. Malignant melanoma cells exhibit increased levels of autophagy, a process of intracellular digestion that allows cells to survive various stresses including chemotherapies, resulting in reduced patient survival. Autophagy can be inhibited by chemicals like chloroquine (CQ), which prevents fusion of autophagosomes to lysosomes, resulting in autophagosome accumulation in most systems. Here, we describe how tested CQ to see whether it could sensitize B16F10 metastatic mouse melanoma cells to the anticancer activities of the natural compounds ursolic acid (UA) and resveratrol (RES). CQ with UA or RES strongly and synergistically reduced the viability of B16F10 mouse melanoma and A375 human melanoma cells. Surprisingly, flow cytometry of acridine orange-stained cells showed that UA or RES in combination with CQ significantly reduced autophagosome levels. Western blotting analysis revealed that CQ plus UA or RES paradoxically increased LC3II, indicative of autophagosome accumulation. In addition, CQ plus RES synergistically decreased the levels of both autophagy initiator beclin-1 and autophagy supporter p62. These results indicate that CQ with UA or RES strongly and synergistically reduces the viability of B16F10 and A375 melanoma cells. However, studies on B16F10 cells have shown that the synergistic effect was not mediated by inhibition of autophagy induced by UA or RES. These compounds are well-tolerated in humans, and CQ has shown promise as an adjuvant therapy. These combinations may be valuable treatment strategies for melanoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Chloroquine/pharmacology , Melanoma, Experimental/drug therapy , Skin Neoplasms/drug therapy , Stilbenes/pharmacology , Triterpenes/pharmacology , Animals , Autophagy/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Humans , Melanoma, Experimental/pathology , Mice , Resveratrol , Signal Transduction/drug effects , Skin Neoplasms/pathology , Ursolic Acid
6.
Mol Carcinog ; 53(9): 753-63, 2014 Sep.
Article in English | MEDLINE | ID: mdl-23625588

ABSTRACT

Glucocorticoids (GCs) are well-known anti-inflammatory compounds, but they also inhibit cell proliferation depending on cell type. Similarly, peroxisome proliferator-activated receptors (PPARα, PPARδ, and PPARγ) also possess anti-proliferation properties beyond their canonical roles as metabolic mediators. In the present study, we investigated the potential additive or synergistic inhibitory effects on cancer cell proliferation by simultaneous application of fenofibrate and budesonide, agonists for PPARα and glucocorticoid receptor, respectively. We observed differential effects on cell proliferation in A549 and SK-MES-1 lung cancer cells by budesonide and fenofibrate. Fenofibrate inhibited cell proliferation in both TP53 wild type and deficient lung cancer cells. The anti-proliferation effect of budesonide in TP53 wild type A549 cells was abolished in SK-MES-1 cells that do not have wild type TP53 protein. An additive effect against cell proliferation by budesonide and fenofibrate combination was observed only in TP53 wild type A549 cancer cells. Analysis of cell cycle distribution and cyclin profile indicated that the inhibition of cell proliferation was associated with G1 cell cycle arrest. The suppression of NF-κB activity and ERK signaling may contribute to the inhibition of cell proliferation by budesonide and or fenofibrate. The additive inhibitory effect on cell proliferation by budesonide and fenofibrate combination suggests that the same or greater therapeutic effect could be achieved with reduced dosage and side effects when the two compounds are applied simultaneously.


Subject(s)
Adenocarcinoma/drug therapy , Budesonide/pharmacology , Cell Proliferation/drug effects , Fenofibrate/pharmacology , Lung Neoplasms/drug therapy , PPAR alpha/agonists , Receptors, Glucocorticoid/agonists , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Anti-Inflammatory Agents/pharmacology , Blotting, Western , Cell Cycle/drug effects , Flow Cytometry , Humans , Hypolipidemic Agents/pharmacology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , PPAR alpha/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Glucocorticoid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
Mol Cancer Res ; 11(12): 1521-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24072817

ABSTRACT

UNLABELLED: Ursolic acid, present in apples, rosemary, and other sources, is known to inhibit tumor formation and tumor cell viability in multiple systems, including skin. However, various cancers are resistant to ursolic acid treatment. Herein, skin carcinoma cells (Ca3/7) as compared with skin papilloma cells (MT1/2) displayed more resistance to ursolic acid-induced cytotoxicity. Interestingly, Ca3/7 cells had elevated levels of P-glycoprotein (P-gp), an ATP-dependent efflux pump that mediates resistance to chemotherapy in preclinical and clinical settings, and not only accumulated less but also more rapidly expelled the P-gp substrate rhodamine 123 (Rh123) indicating ursolic acid is transported by P-gp. To determine whether P-gp inhibition can enhance ursolic acid-mediated cytotoxicity, cells were challenged with P-gp inhibitors verapamil or cyclosporin A. Alternatively, cells were pretreated with the natural compound resveratrol, a known chemotherapy sensitizer. Verapamil and resveratrol enhanced the effects of ursolic acid in both cell lines, whereas cyclosporin A only did so in Ca3/7 cells. Similarly, verapamil inhibited Rh123 efflux in both lines, whereas cyclosporin A only inhibited Rh123 efflux in Ca3/7 cells. Resveratrol did not inhibit Rh123 efflux in either line, indicating the synergistic effects of resveratrol and ursolic acid are not manifest by inhibition of P-gp-mediated efflux of ursolic acid. These results indicate that the anti-skin cancer effects of ursolic acid are enhanced with P-gp inhibitors. In addition, resveratrol and ursolic acid interact synergistically, but not through inhibition of P-gp. IMPLICATIONS: Resveratrol and/or p-glycoprotein inhibitors in combination with ursolic acid are an effective anti-skin cancer regimen.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , Cyclosporine/pharmacology , Skin Neoplasms/drug therapy , Stilbenes/pharmacology , Triterpenes/pharmacology , Verapamil/pharmacology , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Drug Synergism , Mice , Resveratrol , Rhodamine 123/metabolism , Triterpenes/pharmacokinetics , Ursolic Acid
8.
J Cancer ; 4(5): 383-90, 2013.
Article in English | MEDLINE | ID: mdl-23833683

ABSTRACT

Our previous research has shown that plasma fibronectin promotes lung metastasis by facilitating tumor cell invasion in clotted plasma. To evaluate the role of clotted plasma for tumor cell survival, we treated B16F1 cells embedded in a 3-dimensional matrix of fibrin with tumor necrosis factor α (TNFα), a cytokine with anti-tumor activity. Under these conditions, TNFα caused significant cytotoxicity, which was prevented when we added plasma fibronectin to the fibrin clot. Fibronectin-mediated TNFα resistance was dependent on PI3-kinase, which also mediated the pro-adhesive and pro-invasive effects of plasma fibronectin on tumor cells. To further investigate the role of plasma fibronectin in tumor cell signaling, we performed a gene array that showed specific upregulation of Tie2 in B16F1 cells embedded in fibrin-fibronectin compared to fibrin. Importantly, inhibition of Tie2 resulted in decreased tumor cell invasion, reduced colony formation and increased tumor cell death in response to TNFα. Together, our findings indicate that plasma fibronectin induces tumor cell invasion and protects tumor cells from the cytotoxic effects of inflammatory mediators through up-regulation of Tie2.

9.
Mol Cancer Res ; 11(2): 194-203, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23204394

ABSTRACT

High-grade non-muscle-invasive bladder cancer is commonly treated with Bacillus Calmette-Guérin, an immunotherapeutic that depends on fibronectin and tumor cell integrin α5ß1 for internalization into bladder cancer cells. We previously showed that the anti-angiogenic peptide CLT1 forms cytotoxic complexes with fibronectin that are cooperatively internalized into proliferating endothelium through ligation of integrins and chloride intracellular channel 1. While CLT1 has no effect on mature, differentiated cells, we show here that CLT1 is highly cytotoxic for a panel of bladder tumor cell lines as well as a variety of cell lines derived from kidney, lung, breast, and prostate cancer. Paralleling our previous results, we found CLT1-induced tumor cell death to be increased in the presence of fibronectin, which mediated CLT1 internalization and subsequent autophagic cell death in a mechanism that depends on tumor cell integrin α5ß1 and chloride intracellular channel 3 (CLIC3). This mechanistic link was further supported by our results showing upregulation of α5ß1 and CLIC3 in CLT1-responsive tumor cell lines and colocalization with CLT1 in tumor tissues. Incubating tumor tissue from patients with bladder cancer with fluorescein-conjugated CLT1 resulted in a strong and specific fluorescence whereas normal bladder tissue remained negative. On the basis of its affinity for bladder tumor tissue and strong antitumor effects, we propose that CLT1 could be useful for targeting bladder cancer.


Subject(s)
Integrin alpha5beta1/metabolism , Peptides, Cyclic/pharmacology , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism , Autophagy/drug effects , Cell Line, Tumor , Fibronectins/pharmacology , Gene Silencing , Humans , Integrin alpha5beta1/genetics , Molecular Targeted Therapy , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Oligopeptides/pharmacology , Peptides, Cyclic/pharmacokinetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Signal Transduction , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/genetics
10.
Angiogenesis ; 15(1): 115-29, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22203240

ABSTRACT

Angiogenesis is important for tumor growth and metastasis. CLT1 (CGLIIQKNEC), a peptide that binds to tumor interstitial spaces in the presence of fibrin-fibronectin, has structural similarity to the anti-angiogenic ß-sheet peptides anastellin and anginex. This similarity is reflected in the ability of CLT1 to form co-aggregates with fibronectin that induce an unfolded protein response and cause autophagic cell death in proliferating endothelial cells. CLT1 cytotoxicity is mediated at least in parts by a novel CLT1 binding protein, Chloride Intracellular Channel 1 (CLIC1), which promotes internalization of CLT1-fibronectin co-aggregates in a mechanism that depends on the LIIQK amino acid sequence of CLT1. LIIQK encompasses amino acid residues relevant for CLT1 binding to CLIC1 and in addition, facilitates the formation of CLT1-fibronectin co-aggregates, which in turn promote translocation of CLIC1 to the endothelial cell surface through ligation of integrin αvß3. Paralleling the in vitro results, we found that CLT1 co-localizes with CLIC1 and fibronectin in angiogenic blood vessels in vivo, and that CLT1 treatment inhibited angiogenesis and tumor growth. Our findings show that CLT1 is a new anti-angiogenic compound, and its mechanism of action is to form co-aggregates with fibronectin, which bind to angiogenic endothelial cells through integrins, become internalized through CLIC1 and elicit a cytotoxic unfolded protein response. The simple structure and high potency of CLT1 make it a potentially useful compound for anti-angiogenic treatments.


Subject(s)
Chloride Channels/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Fibronectins/metabolism , Neovascularization, Physiologic/drug effects , Peptides, Cyclic/pharmacology , Amino Acid Sequence , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Death/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Endocytosis/drug effects , Fibronectins/chemistry , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Male , Mice , Mice, Nude , Molecular Sequence Data , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Peptides, Cyclic/chemistry , Peptides, Cyclic/therapeutic use , Protein Structure, Quaternary
11.
Cancer Res ; 70(11): 4327-34, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20501851

ABSTRACT

The attachment of circulating tumor cells to the blood vessels of distant organs is an important step in metastasis. We show here that experimental lung metastasis by two cell lines, B16F1 melanoma and 3LL lung carcinoma, is greatly reduced in transgenic mice that lack plasma fibronectin. This multifunctional adhesive glycoprotein becomes cross-linked to fibrin during clotting. Here, we report that eliminating plasma fibronectin from the blood circulation reverses the prometastatic effects of blood clotting and tumor cell integrin alphavbeta3. In vitro studies showed that fibrin-fibronectin complexes, but not purified fibrin, supported tumor cell attachment and invasion. These functions correlate with the ability of fibrin-fibronectin complexes to induce the activation of integrin alphavbeta3. Our findings reveal an important contribution of plasma fibronectin in lung metastasis. Furthermore, they suggest that the previously noted effects of blood clotting on lung metastasis might be mediated in part by a fibronectin-alphavbeta3 integrin axis, in which plasma fibronectin has to be incorporated into the blood clot.


Subject(s)
Carcinoma, Lewis Lung/blood , Carcinoma, Lewis Lung/secondary , Fibronectins/blood , Lung Neoplasms/blood , Lung Neoplasms/secondary , Melanoma, Experimental/blood , Melanoma, Experimental/secondary , Animals , Blood Coagulation/physiology , Carcinoma, Lewis Lung/pathology , Cell Adhesion/physiology , Fibrin/metabolism , Integrin alphaVbeta3/blood , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Invasiveness
12.
Clin Chem ; 54(1): 53-60, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18024530

ABSTRACT

BACKGROUND: The analysis of bodily fluids using SELDI-TOF MS has been reported to identify signatures of spectral peaks that can be used to differentiate patients with a specific disease from normal or control patients. This report is the 2nd of 2 companion articles describing a validation study of a SELDI-TOF MS approach with IMAC surface sample processing to identify prostatic adenocarcinoma. METHODS: We sought to derive a decision algorithm for classification of prostate cancer from SELDI-TOF MS spectral data from a new retrospective sample cohort of 400 specimens. This new cohort was selected to minimize possible confounders identified in the previous study described in the companion paper. RESULTS: The resulting new classifier failed to separate patients with prostate cancer from biopsy-negative controls; nor did it separate patients with prostate cancer with Gleason scores <7 from those with Gleason scores > or =7. CONCLUSIONS: In this, the 2nd stage of our planned validation process, the SELDI-TOF MS-based protein expression profiling approach did not perform well enough to advance to the 3rd (prospective study) stage. We conclude that the results from our previous studies-in which differentiation between prostate cancer and noncancer was demonstrated-are not generalizable. Earlier study samples likely had biases in sample selection that upon removal, as in the present study, resulted in inability of the technique to discriminate cancer from noncancer cases.


Subject(s)
Adenocarcinoma/diagnosis , Biomarkers, Tumor/blood , Blood Specimen Collection , Prostatic Neoplasms/diagnosis , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Aged , Aged, 80 and over , Bias , Case-Control Studies , Humans , Male , Middle Aged , Prospective Studies , Prostate-Specific Antigen/blood , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteomics , Retrospective Studies , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
13.
Clin Chem ; 54(1): 44-52, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17981926

ABSTRACT

BACKGROUND: This report and a companion report describe a validation of the ability of serum proteomic profiling via SELDI-TOF mass spectrometry to detect prostatic cancer. Details of this 3-stage process have been described. This report describes the development of the algorithm and results of the blinded test for stage 1. METHODS: We derived the decision algorithm used in this study from the analysis of serum samples from patients with prostate cancer (n = 181) and benign prostatic hyperplasia (BPH) (n = 143) and normal controls (n = 220). We also derived a validation test set from a separate, geographically diverse set of serum samples from 42 prostate cancer patients and 42 controls without prostate cancer. Aliquots were subjected to randomization and blinded analysis, and data from each laboratory site were subjected to the decision algorithm and decoded. RESULTS: Using the data collected from the validation test set, the decision algorithm was unsuccessful in separating cancer from controls with any predictive utility. Analysis of the experimental data revealed potential sources of bias. CONCLUSION: The ability of the decision algorithm to successfully differentiate between prostate cancer, BPH, and control samples using data derived from serum protein profiling was compromised by bias.


Subject(s)
Biomarkers, Tumor/blood , Blood Specimen Collection , Prostatic Neoplasms/diagnosis , Algorithms , Bias , Decision Support Techniques , Humans , Male , Middle Aged , Predictive Value of Tests , Prostatic Hyperplasia/diagnosis , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/metabolism , Proteomics , ROC Curve , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
14.
Proc Natl Acad Sci U S A ; 104(41): 16056-61, 2007 Oct 09.
Article in English | MEDLINE | ID: mdl-17911243

ABSTRACT

Heat-labile enterotoxin (LT) from enterotoxigenic Escherichia coli is a heterohexameric protein consisting of an enzymatically active A subunit, LTA, and a carrier pentameric B subunit, LTB. It is clear from the crystal structure of LTB that the N-terminal alpha1 helix lies outside the core structure. However, the function of the N-terminal alpha1 helix of LTB is unknown. The present work was carried out to investigate the effect of site-directed mutagenesis of the alpha1 helix on LTB synthesis. Six amino acids (PQSITE) located at positions 2-7 from the N terminus, including 4 aa from the alpha1 helix, were deleted by site-directed mutagenesis. The deletion resulted in complete inhibition of LTB expression in E. coli when expressed along with its signal sequence. A single amino acid deletion within the alpha1 helix also resulted in loss of expression. However, a single amino acid deletion outside the alpha1 helix did not affect LTB synthesis. Mutant proteins, whose synthesis was not detected in vivo, could be successfully translated in vitro by using the coupled transcription-translation system. Immunoblot analysis, Northern blot analysis, and in vitro transcription-translation data collectively indicate that the lack of synthesis of the mutant proteins is caused by the immediate degradation of the expressed product by cellular proteases rather than by faulty translation of mutant LTB mRNA. Coexpression of the LTA could not rescue the degradation of LTB mutants.


Subject(s)
Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Enterotoxigenic Escherichia coli/genetics , Enterotoxigenic Escherichia coli/metabolism , Enterotoxins/genetics , Enterotoxins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Amino Acid Sequence , Bacterial Toxins/chemistry , Base Sequence , DNA Primers/genetics , Enterotoxins/chemistry , Escherichia coli Proteins/chemistry , Genes, Bacterial , Magnesium/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Protein Structure, Quaternary , Protein Structure, Secondary , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Deletion , Transcription, Genetic
15.
Prostate ; 67(14): 1565-75, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17705230

ABSTRACT

BACKGROUND: We recently demonstrated the protein expression profiling of Dunning rat tumor cell lines of varying metastatic potential (G (0%), AT-1 ( approximately 20%), and MLL (100%)) using SELDI-TOF-MS. As a parallel effort, we have been pursuing the identification of the protein(s) comprising the individual discriminatory "peaks" and evaluating their utility as potential biomarkers for prostate cancer progression. METHODS: To identify the observed SELDI-TOF-MS m/z (mass/charge) values with discriminatory expression between different sublines, we employed a combination of chemical pre-fractionation, liquid chromatography, gel electrophoresis and tandem mass spectroscopy. Identified proteins were then verified by immuno-assay and Western analysis. RESULTS: A 17.5 K m/z SELDI-TOF-MS peak was found to retain discriminatory value in each of two separate study-sets with an increased expression in the metastatic MLL line. Sequence identification and subsequent immunoassays verified that Histone H2B is the observed 17.5 K m/z SELDI peak. SELDI-based immuno-assay and Western Blotting revealed that Histone H2B is specifically over-expressed in metastatic MLL lines. CONCLUSIONS: SELDI-TOF MS analysis of the Dunning prostate cancer cell lines confirmed the consistent overexpression of a 17.5 K m/z peak in metastatic MLL subline. The 17.5 kDa protein from MLL has been isolated and identified as Histone H2B.


Subject(s)
Adenocarcinoma/metabolism , Biomarkers, Tumor/metabolism , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Protein Array Analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Adenocarcinoma/chemistry , Adenocarcinoma/secondary , Animals , Biomarkers, Tumor/analysis , Cell Line, Tumor , Chromatography, High Pressure Liquid , Disease Progression , Electrophoresis, Polyacrylamide Gel , Male , Neoplasm Metastasis , Neoplasm Proteins/analysis , Prostatic Neoplasms/chemistry , Prostatic Neoplasms/pathology , Proteomics , Rats , Tandem Mass Spectrometry
16.
FEMS Microbiol Lett ; 263(2): 229-35, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16978361

ABSTRACT

Triosephosphate isomerase (TPI), one of the key enzymes of the glycolytic pathway, is an attractive drug target against Mycobacterium tuberculosis as glycolysis provides the majority of the organism's energy requirements inside macrophages. To carry out biochemical and biophysical characterization, purified recombinant M. tuberculosis TPI produced in Escherichia coli was used. Mass spectrum analysis showed M. tuberculosis rTPI to be of 28 213 Da. The biologically active enzyme is a homodimer as determined by gel filtration chromatography. The M. tuberculosis TPI had a pH optimum in the range of 6-8 and a temperature optimum around 37 degrees C. Circular dichroism spectra analysis revealed that loss of secondary structure of rTPI occurs around 60 degrees C. Metal cations were not required for M. tuberculosis TPI activity. The k(cat) was 4.1 x 10(6) min(-1). Importantly, the apparent K(m) value of M. tuberculosis rTPI for the substrate glyceraldehyde-3-phosphate is 84 microM which is sevenfold higher than the value reported for human TPI. The difference in K(m) is indicative of the difference in the active site of the human and M. tuberculosis TPI, which can be exploited for drug designing specifically targeting M. tuberculosis TPI.


Subject(s)
Mycobacterium tuberculosis/enzymology , Triose-Phosphate Isomerase/analysis , Triose-Phosphate Isomerase/metabolism , DNA, Bacterial/analysis , Genes, Bacterial , Mycobacterium tuberculosis/classification , Mycobacterium tuberculosis/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
17.
Mol Cell Proteomics ; 5(10): 1957-67, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16760258

ABSTRACT

The application of mass spectrometry to identify disease biomarkers in clinical fluids like serum using high throughput protein expression profiling continues to evolve as technology development, clinical study design, and bioinformatics improve. Previous protein expression profiling studies have offered needed insight into issues of technical reproducibility, instrument calibration, sample preparation, study design, and supervised bioinformatic data analysis. In this overview, new strategies to increase the utility of protein expression profiling for clinical biomarker assay development are discussed with an emphasis on utilizing differential lectin-based glycoprotein capture and targeted immunoassays. The carbohydrate binding specificities of different lectins offer a biological affinity approach that complements existing mass spectrometer capabilities and retains automated throughput options. Specific examples using serum samples from prostate cancer and hepatocellular carcinoma subjects are provided along with suggested experimental strategies for integration of lectin-based methods into clinical fluid expression profiling strategies. Our example workflow incorporates the necessity of early validation in biomarker discovery using an immunoaffinity-based targeted analytical approach that integrates well with upstream discovery technologies.


Subject(s)
Glycoproteins/blood , Lectins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Amino Acid Sequence , Biomarkers/blood , Biomarkers/chemistry , Glycoproteins/chemistry , Humans , Molecular Sequence Data , Proteomics
18.
J Cell Biochem ; 98(3): 496-503, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16552720

ABSTRACT

There has been an impressive emergence of mass spectrometry based technologies applied toward the study of proteins. Equally notable is the rapid adaptation of these technologies to biomedical approaches in the realm of clinical proteomics. Concerted efforts toward the elucidation of the proteomes of organ sites or specific disease state are proliferating and from these efforts come the promise of better diagnostics/prognostics and therapeutic intervention. Prostate cancer has been a focus of many such studies with the promise of improved care to patients via biomarkers derived from these proteomic approaches. The newer technologies provide higher analytical capabilities, employ automated liquid handling systems, fractionation techniques and bioinformatics tools for greater sensitivity and resolving power, more robust and higher throughput sample processing, and greater confidence in analytical results. In this prospects, we summarize the proteomic technologies applied to date in prostate cancer, along with their respective advantages and disadvantages. The development of newer proteomic strategies for use in future applications is also discussed.


Subject(s)
Biomarkers, Tumor/analysis , Biomarkers, Tumor/chemistry , Prostatic Neoplasms/chemistry , Prostatic Neoplasms/diagnosis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Humans , Male , Proteomics
19.
Clin Cancer Res ; 11(3): 1073-85, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15709174

ABSTRACT

PURPOSE: We recently showed that protein expression profiling of serum using surface-enhanced laser desorption/ionization time-of-flight mass spectrometry (SELDI-TOF MS) has potential as a diagnostic approach for detection of prostate cancer. As a parallel effort, we have been pursuing the identification of the protein(s) comprising the individual discriminatory "peaks" and evaluating their utility as potential biomarkers for prostate disease. EXPERIMENTAL DESIGN: We employed liquid chromatography, gel electrophoresis and tandem mass spectroscopy to isolate and identify a protein that correlates with observed SELDI-TOF MS mass/charge (m/z) values. Immunodepletion, immunoassay, and Western analysis were used to verify that the identified protein generated the observed SELDI peak. Subsequent immunohistochemistry was used to examine the expression of the proteins in prostate tumors. RESULTS: An 8,946 m/z SELDI-TOF MS peak was found to retain discriminatory value in each of two separate data sets with an increased expression in the diseased state. Sequence identification by liquid chromatography-MS/MS and subsequent immunoassays verified that an isoform of apolipoprotein A-II (ApoA-II) is the observed 8,946 m/z SELDI peak. Immunohistochemistry revealed that ApoA-II is overexpressed in prostate tumors. SELDI-based immunoassay revealed that an 8.9-kDa isoform of ApoA-II is specifically overexpressed in serum from individuals with prostate cancer. ApoA-II was also overexpressed in the serum of individuals with prostate cancer who have normal prostate-specific antigen (0-4.0 ng/mL). CONCLUSIONS: We have identified an isoform of ApoA-II giving rise to an 8.9K m/z SELDI "peak" that is specifically overexpressed in prostate disease. The ability of ApoA-II to detect disease in patients with normal prostate-specific antigen suggests potential utility of the marker in identifying indolent disease.


Subject(s)
Apolipoprotein A-II/blood , Biomarkers, Tumor/blood , Prostatic Neoplasms/blood , Apolipoprotein A-II/analysis , Apolipoprotein A-II/isolation & purification , Biomarkers, Tumor/analysis , Humans , Immunohistochemistry , Male , Prostate/chemistry , Prostate/pathology , Prostate-Specific Antigen/blood , Prostatic Intraepithelial Neoplasia/blood , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/metabolism , Protein Isoforms/analysis , Protein Isoforms/blood , Protein Isoforms/isolation & purification , Sensitivity and Specificity , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
20.
Cancer Inform ; 1: 86-97, 2005.
Article in English | MEDLINE | ID: mdl-19305634

ABSTRACT

Multiple studies have reported that surface enhanced laser desorption/ionization time of flight mass spectroscopy (SELDI-TOF-MS) is useful in the early detection of disease based on the analysis of bodily fluids. Use of any multiplex mass spectroscopy based approach as in the analysis of bodily fluids to detect disease must be analyzed with great care due to the susceptibility of multiplex and mass spectroscopy methods to biases introduced via experimental design, patient samples, and/or methodology. Specific biases include those related to experimental design, patients, samples, protein chips, chip reader and spectral analysis. Contributions to biases based on patients include demographics (e.g., age, race, ethnicity, sex), homeostasis (e.g., fasting, medications, stress, time of sampling), and site of analysis (hospital, clinic, other). Biases in samples include conditions of sampling (type of sample container, time of processing, time to storage), conditions of storage, (time and temperature of storage), and prior sample manipulation (freeze thaw cycles). Also, there are many potential biases in methodology which can be avoided by careful experimental design including ensuring that cases and controls are analyzed randomly. All the above forms of biases affect any system based on analyzing multiple analytes and especially all mass spectroscopy based methods, not just SELDI-TOF-MS. Also, all current mass spectroscopy systems have relatively low sensitivity compared with immunoassays (e.g., ELISA). There are several problems which may be unique to the SELDI-TOF-MS system marketed by Ciphergen(®). Of these, the most important is a relatively low resolution (±0.2%) of the bundled mass spectrometer which may cause problems with analysis of data. Foremost, this low resolution results in difficulties in determining what constitutes a "peak" if a peak matching approach is used in analysis. Also, once peaks are selected, the peaks may represent multiple proteins. In addition, because peaks may vary slightly in location due to instrumental drift, long term identification of the same peaks may prove to be a challenge. Finally, the Ciphergen(®) system has some "noise" of the baseline which results from the accumulation of charge in the detector system. Thus, we must be very aware of the factors that may affect the use of proteomics in the early detection of disease, in determining aggressive subsets of cancers, in risk assessment and in monitoring the effectiveness of novel therapies.

SELECTION OF CITATIONS
SEARCH DETAIL
...