Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.365
Filter
1.
Acta Neuropathol Commun ; 9(1): 154, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34530925

ABSTRACT

Brain accumulation of amyloid-beta (Aß) is a crucial feature in Alzheimer´s disease (AD) and cerebral amyloid angiopathy (CAA), although the pathophysiological relationship between these diseases remains unclear. Numerous proteins are associated with Aß deposited in parenchymal plaques and/or cerebral vessels. We hypothesized that the study of these proteins would increase our understanding of the overlap and biological differences between these two pathologies and may yield new diagnostic tools and specific therapeutic targets. We used a laser capture microdissection approach combined with mass spectrometry in the APP23 transgenic mouse model of cerebral-ß-amyloidosis to specifically identify vascular Aß-associated proteins. We focused on one of the main proteins detected in the Aß-affected cerebrovasculature: MFG-E8 (milk fat globule-EGF factor 8), also known as lactadherin. We first validated the presence of MFG-E8 in mouse and human brains. Immunofluorescence and immunoblotting studies revealed that MFG-E8 brain levels were higher in APP23 mice than in WT mice. Furthermore, MFG-E8 was strongly detected in Aß-positive vessels in human postmortem CAA brains, whereas MFG-E8 was not present in parenchymal Aß deposits. Levels of MFG-E8 were additionally analysed in serum and cerebrospinal fluid (CSF) from patients diagnosed with CAA, patients with AD and control subjects. Whereas no differences were found in MFG-E8 serum levels between groups, MFG-E8 concentration was significantly lower in the CSF of CAA patients compared to controls and AD patients. Finally, in human vascular smooth muscle cells MFG-E8 was protective against the toxic effects of the treatment with the Aß40 peptide containing the Dutch mutation. In summary, our study shows that MFG-E8 is highly associated with CAA pathology and highlights MFG-E8 as a new CSF biomarker that could potentially be used to differentiate cerebrovascular Aß pathology from parenchymal Aß deposition.


Subject(s)
Antigens, Surface/biosynthesis , Brain/metabolism , Brain/pathology , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/pathology , Milk Proteins/biosynthesis , Aged , Animals , Antigens, Surface/genetics , Biomarkers/metabolism , Cells, Cultured , Cerebral Amyloid Angiopathy/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Milk Proteins/genetics
2.
Sci Rep ; 11(1): 9777, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33963223

ABSTRACT

Bladder cancer (BCa) patients are diagnosed by cytology and cystoscopy. However, these diagnostic tests bear some limitations. We sought for reliable biomarkers to better determine BCa extension. Prostate-specific membrane antigen (PSMA) appears to fulfill this requirement in prostate cancer but its role in BCa has not been established yet. We then analyzed 87 bladder tissue samples from 74 patients assessing PSMA expression by immunohistochemistry. The median PSMA expression, exclusively found in tumor neovasculature, in terms of H-score significantly differed between non-tumor samples and tumor samples (p = 0.00288) showing a higher neovasculature-related PSMA expression. No differences were observed in relation to tumor type, grade and stage. BCa neovasculature-related PSMA overexpression may be useful in defining the degree of extension of the neoplasm. In addition, testing PSMA expression by immunohistochemistry may hold theranostic implications both considering anti-angiogenesis agents and radio-labelled PSMA ligands for intracavitary radionuclide therapy. In our opinion, BCa neovasculature-related PSMA overexpression may be considered an apt target for anti-angiogenesis and radionuclide treatment in BCa, once the evaluation of tumor-retention time for the appropriateness of long half-life therapeutic PSMA ligands as radionuclide treatment will be performed.


Subject(s)
Antigens, Surface/biosynthesis , Biomarkers, Tumor/biosynthesis , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glutamate Carboxypeptidase II/biosynthesis , Neoplasm Proteins/biosynthesis , Urinary Bladder Neoplasms , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Retrospective Studies , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
3.
PLoS Negl Trop Dis ; 15(3): e0008352, 2021 03.
Article in English | MEDLINE | ID: mdl-33760809

ABSTRACT

Leishmania parasites cycle between sand-fly vectors and mammalian hosts adapting to alternating environments by stage-differentiation accompanied by changes in the proteome profiles. Translation regulation plays a central role in driving the differential program of gene expression since control of gene regulation in Leishmania is mostly post-transcriptional. The Leishmania genome encodes six eIF4E paralogs, some of which bind a dedicated eIF4G candidate, and each eIF4E is assumed to have specific functions with perhaps some overlaps. However, LeishIF4E2 does not bind any known eIF4G ortholog and was previously shown to comigrate with the polysomal fractions of sucrose gradients in contrast to the other initiation factors that usually comigrate with pre-initiation and initiation complexes. Here we deleted one of the two LeishIF4E2 gene copies using the CRISPR-Cas9 methodology. The deletion caused severe alterations in the morphology of the mutant cells that became round, small, and equipped with a very short flagellum that did not protrude from its pocket. Reduced expression of LeishIF4E2 had no global effect on translation and growth, unlike other LeishIF4Es; however, there was a change in the proteome profile of the LeishIF4E2(+/-) cells. Upregulated proteins were related mainly to general metabolic processes including enzymes involved in fatty acid metabolism, DNA repair and replication, signaling, and cellular motor activity. The downregulated proteins included flagellar rod and cytoskeletal proteins, as well as surface antigens involved in virulence. Moreover, the LeishIF4E2(+/-) cells were impaired in their ability to infect cultured macrophages. Overall, LeishIF4E2 does not behave like a general translation factor and its function remains elusive. Our results also suggest that the individual LeishIF4Es perform unique functions.


Subject(s)
Adaptation, Physiological/genetics , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Leishmania/genetics , Amino Acid Sequence/genetics , Animals , Antigens, Surface/biosynthesis , Antigens, Surface/genetics , CRISPR-Cas Systems/genetics , Cells, Cultured , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , Gene Expression Regulation/genetics , Humans , Macrophages/parasitology , Psychodidae/parasitology , Sequence Alignment
4.
Int J Neurosci ; 131(1): 15-24, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32098538

ABSTRACT

Background: Ischemic stroke is a complex pathological process, involving inflammatory reaction, energy metabolism disorder, free radical injury, cell apoptosis and other aspects. Accumulating evidences have revealed that MFG-E8 had a protective effect on multiple organ injuries. However, the comprehensive function and mechanism of MFG-E8 in ischemic brain remain largely unclear.Methods: BV-2 cells were treated with recombinant murine MFG-E8 (rmMFG-E8) or/and Colivelin TFA after exposing for 4 h with oxygen glucose deprivation (OGD). Cell viability and apoptosis were assessed by MTT assay and Flow cytometry. RT-qPCR and Western blot assays were applied to examine the expression levels of MFG-E8, apoptosis-related proteins and M1/M2 polarization markers.Results: Our results demonstrated that OGD significantly inhibited microglial viability and facilitated apoptosis. In addition, we found that OGD downregulated MFG-E8 expression, and MFG-E8 inhibited OGD-induced microglial apoptosis and promoted microglial M2 polarization. In terms of mechanism, we proved that MFG-E8 regulated OGD-induced microglial M1/M2 polarization by inhibiting p-STAT3 and SOCS3 expressions, which was reversed by STAT3 activator (Colivelin TFA). Finally, we verified MFG-E8 alleviated OGD-induced neuronal cell apoptosis by M2 polarization of BV-2 cells.Conclusions: We demonstrated that MFG-E8 reduced neuronal cell apoptosis by enhancing activation of microglia via STAT3 signaling. Therefore, we suggested that MFG-E8 might provide a novel mechanism for ischemic stroke.


Subject(s)
Antigens, Surface/biosynthesis , Cell Hypoxia/physiology , Glucose/deficiency , Microglia/metabolism , Milk Proteins/biosynthesis , Neurons/metabolism , STAT3 Transcription Factor/biosynthesis , Animals , Apoptosis/physiology , Cell Line , Cell Polarity/physiology , Coculture Techniques , Mice , Milk Proteins/antagonists & inhibitors
5.
Toxicol Lett ; 322: 50-57, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-31958493

ABSTRACT

Allergic contact dermatitis (ACD) is an important occupational and environmental disease caused by topical exposure to chemical allergens. In the EU, it has been calculated that 4 % of animals are used in toxicity test for the assessment of skin sensitization (Peiser et al., 2012). To come a complete replacement of animals, evaluation of relative skin sensitization potency is necessary. The identification of mechanisms influencing allergen potency requires a better understanding of molecular events that trigger cell activation. Therefore, (i) the effects of selected allergens on surface markers expression and cytokines release in contact allergen-induced cell activation were assessed, and (ii) the role of Protein Kinase C (PKC) beta activation in contact allergen-induced cell activation was investigated. The human pro-myelocytic cell line THP-1 was used as experimental model surrogate of dendritic cells. Cells were exposed to select contact allergens of different potency and cell surface marker expression (CD80, CD86, HLA-DR) was determined by flow cytometry analysis. Cytokines production (IL-6, IL-8, IL-10, IL-12p40, IL-18) was evaluated with specific sandwich ELISA. The effective contribution of PKC beta in chemical allergen-induced cell activation was assessed by Western Blot analysis (PKC beta activation) and using a specific PKC beta inhibitor (PKC beta pseudosubstrate). In addition, to investigate if contact allergens are able to induce indeed dendritic cells (DCs) maturation, THP-1 cells were differentiated to immature DC and then exposed to contact allergen of different potency. Overall, our finding provides insights into the process of sensitization and strength of cell activation associated with allergens of different potency. Results obtained suggest that contact allergens of different potency are able to induce a different degree of activation of dendritic cells maturation involved in the process of ACD.


Subject(s)
Allergens/classification , Animal Testing Alternatives , Dendritic Cells/drug effects , Dermatitis, Allergic Contact , Skin/drug effects , Xenobiotics/classification , Allergens/toxicity , Antigens, Surface/biosynthesis , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , Cytokines/metabolism , Dendritic Cells/enzymology , Dendritic Cells/immunology , Dermatitis, Allergic Contact/enzymology , Dermatitis, Allergic Contact/immunology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Humans , Protein Kinase C beta/metabolism , Skin/enzymology , Skin/immunology , Xenobiotics/toxicity
6.
Pathol Oncol Res ; 26(2): 1263-1268, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31256324

ABSTRACT

Milk fat globule-epidermal growth factor 8 (MFG-E8) is a glycoprotein secreted by the activated macrophages and acts as a bridge between apoptotic cells and phagocytes. Aside from macrophages, a variety of malignant cells also express MFG-E8. The objective of this study is to elucidate the clinical relevance and significance of MFG-E8 in the tumor microenvironment (TME) of patients with oral squamous cell carcinoma (OSCC). We investigated MFG-E8 expression in 74 patients with OSCC by immunohistochemistry and evaluated the relationship between MFG-E8 expression and various clinicopathological factors including immune cell infiltration. MFG-E8 expression was detected in 34 of 74 (45.9%) patients with OSCC and a significant correlation was observed with levels of infiltrating T cells, macrophages, and immunosuppressive M2 macrophages. Furthermore, MFG-E8 expression was also associated with clinical stage, lymphatic/vascular invasion, and Ki-67+ tumor cells but not with survival. Our results suggest that MFG-E8 may play an important role in shaping the immune suppressive network in TME as well as tumor progression.


Subject(s)
Antigens, Surface/biosynthesis , Head and Neck Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Milk Proteins/biosynthesis , Squamous Cell Carcinoma of Head and Neck/immunology , Tumor Microenvironment/immunology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Female , Head and Neck Neoplasms/pathology , Humans , Male , Middle Aged , Squamous Cell Carcinoma of Head and Neck/pathology
7.
Eur J Nucl Med Mol Imaging ; 47(1): 9-15, 2020 01.
Article in English | MEDLINE | ID: mdl-31654093
8.
Macromol Biosci ; 19(10): e1900183, 2019 10.
Article in English | MEDLINE | ID: mdl-31507074

ABSTRACT

Reactive oxygen species (ROS) forming enzymes are of significant interest as anticancer agents due to their potent cytotoxicity. A key challenge in their clinical translation is attaining site-specific delivery and minimizing biodistribution to healthy tissues. Here, complexes composed of the ROS enzyme glucose oxidase (GOX), poly-l-lysine-grafted-polyethylene glycol (PLL-g-PEG), and anti-prostate specific membrane antigen (anti-PSMA) monoclonal antibody are synthesized for localized delivery and uptake in prostate cancer cells. Formation of anti-PSMA-PLL-g-PEG/GOX results in nanoscale complexes ≈30 nm in diameter with a ζ-potential of 6 mV. The anti-PSMA-PLL-g-PEG/GOX complexes show significant cytotoxicity (≈60% reduction in cell viability) against PSMA-expressing LNCaP cells compared to unmodified GOX. Importantly, cytotoxicity in LNCaP cells occurrs concurrently with anti-PSMA-PLL-g-PEG/GOX uptake and increases in intracellular generation of ROS. These results demonstrate that cytotoxicity of ROS inducing enzymes can be enhanced by intracellular delivery compared to equivalent concentrations of free enzyme, providing a novel means for cancer therapy.


Subject(s)
Antigens, Surface/biosynthesis , Aspergillus niger/enzymology , Cytotoxins/pharmacology , Fungal Proteins/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glucose Oxidase/pharmacology , Glutamate Carboxypeptidase II/biosynthesis , Neoplasm Proteins/biosynthesis , Prostatic Neoplasms/drug therapy , Humans , Male , PC-3 Cells , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology
9.
Cir Cir ; 87(S1): 8-16, 2019.
Article in English | MEDLINE | ID: mdl-31501623

ABSTRACT

BACKGROUND: Procedures to remove adiposities and skin, such as dermolipectomy, can develop wounds that are difficult to heal by conventional therapies. Mesenchymal stem cells are indicated as potential candidates for regenerative therapy in wounds, due to their multipotentiality, low immunogenicity, modulating capacity of inflammation and tissue modeling processes. CASE REPORT: Patient with dehiscent chronic ulcer secondary to dermolipectomy, who received cutaneous treatment with mesenchymal stem cells. The therapy induced scar formation and neovascularization, as well as the decrease of infiltrated leukocytes and proinflammatory cytokines. Mesenchymal cells are proposed as an interesting alternative for the treatment of postoperative lesions.


INTRODUCCIÓN: Los procedimientos para retirar adiposidades y piel, como la dermolipectomía, pueden desarrollar heridas difíciles de sanar mediante tratamientos convencionales. Se ha señalado que es posible utilizar las células madre mesenquimales en el tratamiento regenerativo en heridas, en virtud de su multipotencialidad, baja inmunogenicidad, capacidad moduladora de inflamación y procesos modeladores de tejidos. CASO CLÍNICO: Paciente con dehiscencia en úlcera crónica secundaria a dermolipectomía, sometida a tratamiento cutáneo con células madre mesenquimales. Se indujo formación de cicatriz y neovascularización, así como la disminución de leucocitos infiltrados y citocinas proinflamatorias. Se propone a las células mesenquimales como una alternativa interesante para el tratamiento de lesiones postoperatorias.


Subject(s)
Body Contouring/adverse effects , Lipectomy/adverse effects , Mesenchymal Stem Cell Transplantation , Regenerative Medicine/methods , Skin Ulcer/therapy , Surgical Wound Dehiscence/therapy , Wharton Jelly/cytology , Adipogenesis , Adult , Antigens, Surface/biosynthesis , Antigens, Surface/genetics , Cell Separation , Chronic Disease , Cicatrix/etiology , Female , Gene Expression , Humans , Inflammation , Mesenchymal Stem Cells , Neovascularization, Physiologic , Osteogenesis , Skin Ulcer/etiology , Surgical Wound Dehiscence/etiology , Wound Healing
10.
Eur Urol Oncol ; 2(6): 670-676, 2019 11.
Article in English | MEDLINE | ID: mdl-31412006

ABSTRACT

BACKGROUND: Prostate-specific membrane antigen (PSMA) is overexpressed in metastatic castration-resistant prostate cancer (mCRPC) and represents a target for imaging and therapy. We undertook a prospective trial of 177Lu-PSMA-617 radioligand therapy in men with high PSMA expression who progressed after standard therapies. OBJECTIVE: To determine outcomes for men screened for the trial but not treated because of low PSMA expression. DESIGN, SETTING, AND PARTICIPANTS: Patients screened with 68Ga-PSMA-11 and 18F-fluorodeoxyglucose (FDG) positron emission tomography/computed tomography in a prospective trial. Patients ineligible for enrolment with low PSMA expression or FDG-positive PSMA-negative (discordant FDG-avid) disease were assessed. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Subsequent treatments received were recorded. Kaplan-Meier analysis was used to determine overall survival from date of screening. RESULTS AND LIMITATIONS: Sixteen patients (24%) had low PSMA expression (n=8) or discordant FDG-avid disease (n=8). The median prostate-specific antigen doubling-time was 2.1mo. Eleven patients had Gleason ≥8 disease. All patients had previously progressed after docetaxel, 44% after cabazitaxel, and 94% after abiraterone and/or enzalutamide. Nine patients had subsequent systemic antitumour treatment. Fifteen patients died, with median OS of 2.5mo (95% confidence interval 1.7-5.0). Study limitations include uncertainty for imaging thresholds that define low PSMA expression. It is also possible that theranostic therapy could have improved survival in this cohort. CONCLUSIONS: Low PSMA expression or discordant FDG-avid disease in patients with mCRPC who progress after conventional therapies identifies a group with poor prognosis and short survival. PATIENT SUMMARY: The 177Lu-PSMA-617 radioligand may be an effective therapy for patients with advanced prostate cancer who progress after standard therapies. In this report we looked at outcomes for patients who were not eligible for this novel therapy on the basis of low prostate-specific membrane antigen uptake on screening positron emission tomography scans. We found that their outcomes were poor, with short survival.


Subject(s)
Dipeptides/therapeutic use , Heterocyclic Compounds, 1-Ring/therapeutic use , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Radiopharmaceuticals/therapeutic use , Aged , Aged, 80 and over , Antigens, Surface/biosynthesis , Disease-Free Survival , Glutamate Carboxypeptidase II/biosynthesis , Humans , Lutetium , Male , Middle Aged , Prognosis , Prostate-Specific Antigen , Prostatic Neoplasms, Castration-Resistant/mortality , Treatment Outcome
11.
Eur Urol ; 76(4): 469-478, 2019 10.
Article in English | MEDLINE | ID: mdl-31345636

ABSTRACT

BACKGROUND: Prostate-specific membrane antigen (PSMA; folate hydrolase) prostate cancer (PC) expression has theranostic utility. OBJECTIVE: To elucidate PC PSMA expression and associate this with defective DNA damage repair (DDR). DESIGN, SETTING, AND PARTICIPANTS: Membranous PSMA (mPSMA) expression was scored immunohistochemically from metastatic castration-resistant PC (mCRPC) and matching, same-patient, diagnostic biopsies, and correlated with next-generation sequencing (NGS) and clinical outcome data. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Expression of mPSMA was quantitated by modified H-score. Patient DNA was tested by NGS. Gene expression and activity scores were determined from mCRPC transcriptomes. Statistical correlations utilised Wilcoxon signed rank tests, survival was estimated by Kaplan-Meier test, and sample heterogeneity was quantified by Shannon's diversity index. RESULTS AND LIMITATIONS: Expression of mPSMA at diagnosis was associated with higher Gleason grade (p=0.04) and worse overall survival (p=0.006). Overall, mPSMA expression levels increased at mCRPC (median H-score [interquartile range]: castration-sensitive prostate cancer [CSPC] 17.5 [0.0-60.0] vs mCRPC 55.0 [2.8-117.5]). Surprisingly, 42% (n=16) of CSPC and 27% (n=16) of mCRPC tissues sampled had no detectable mPSMA (H-score <10). Marked intratumour heterogeneity of mPSMA expression, with foci containing no detectable PSMA, was observed in all mPSMA expressing CSPC (100%) and 37 (84%) mCRPC biopsies. Heterogeneous intrapatient mPSMA expression between metastases was also observed, with the lowest expression in liver metastases. Tumours with DDR had higher mPSMA expression (p=0.016; 87.5 [25.0-247.5] vs 20 [0.3-98.8]; difference in medians 60 [5.0-95.0]); validation cohort studies confirmed higher mPSMA expression in patients with deleterious aberrations in BRCA2 (p<0.001; median H-score: 300 [165-300]; difference in medians 195.0 [100.0-270.0]) and ATM (p=0.005; 212.5 [136.3-300]; difference in medians 140.0 [55.0-200]) than in molecularly unselected mCRPC biopsies (55.0 [2.75-117.5]). Validation studies using mCRPC transcriptomes corroborated these findings, also indicating that SOX2 high tumours have low PSMA expression. CONCLUSIONS: Membranous PSMA expression is upregulated in some but not all PCs, with mPSMA expression demonstrating marked inter- and intrapatient heterogeneity. DDR aberrations are associated with higher mPSMA expression and merit further evaluation as predictive biomarkers of response for PSMA-targeted therapies in larger, prospective cohorts. PATIENT SUMMARY: Through analysis of prostate cancer samples, we report that the presence of prostate-specific membrane antigen (PSMA) is extremely variable both within one patient and between different patients. This may limit the usefulness of PSMA scans and PSMA-targeted therapies. We show for the first time that prostate cancers with defective DNA repair produce more PSMA and so may respond better to PSMA-targeting treatments.


Subject(s)
Antigens, Surface/biosynthesis , DNA Repair , Glutamate Carboxypeptidase II/biosynthesis , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Antigens, Surface/analysis , Glutamate Carboxypeptidase II/analysis , Humans , Male , Prostatic Neoplasms, Castration-Resistant/chemistry , Retrospective Studies
12.
Prostate ; 79(12): 1450-1456, 2019 09.
Article in English | MEDLINE | ID: mdl-31233227

ABSTRACT

BACKGROUND: Prostate-specific membrane antigen (PSMA)-based imaging and therapy are increasingly used in the management of prostate cancer. However, low PSMA surface expression in certain patients is a limitation for PSMA-based technologies. We have previously shown that high doses of dutasteride, a 5α-reductase inhibitor generally used for the treatment of benign prostatic enlargement, increase the PSMA expression in vitro. We now further analyzed the concentration- and time-dependent effects of dutasteride in LNCaP cells. METHODS: Androgen receptor (AR) expressing prostate cancer cells (LNCaP) were treated for 7 to 14 days with vehicle control (0.1% dimethyl sulfoxide) or different concentrations of dutasteride (0.25 , 0.5 , 1 , and 5 µM). In addition to cell proliferation, PSMA surface expression was assessed using flow cytometry (FACS) and immunocytochemistry. Total PSMA and AR expression was analyzed by capillary western immunoassay (WES). In addition, tumor cell uptake and internalization assays of 177 Lu-PSMA-617 were performed. RESULTS: Dutasteride treatment resulted in a significant upregulation of PSMA surface expression compared to vehicle control after 7 days in all tested concentrations. After 14 days a further, concentration-dependent increase of PSMA surface expression was detectable. Total PSMA protein expression significantly increased after treatment of cells with high concentrations of dutasteride using 5 µM for 7 or 14 days. However, when lower concentrations were used total PSMA expression was not significantly altered compared to vehicle control. Further testing revealed a dose-dependent increase in uptake and internalization of 177Lu -PSMA-617 after 7 and 14 days. Though, a significantly increased uptake was only observed using a 5 µM dutasteride concentration for 7 days as well as 1 and 5 µM for 14 days. CONCLUSION: Our investigations revealed a concentration- and time-dependent effect of dutasteride on PSMA expression and uptake of 177Lu -PSMA-617 in LNCaP cells. A short-term treatment of patients with high doses of dutasteride might increase the detection rate of PSMA-based imaging and increase the effect of 177Lu -PSMA-617 therapy via upregulation of PSMA expression.


Subject(s)
5-alpha Reductase Inhibitors/pharmacology , Antigens, Surface/biosynthesis , Dutasteride/pharmacology , Glutamate Carboxypeptidase II/biosynthesis , Prostate/drug effects , Prostatic Neoplasms/metabolism , Cell Line, Tumor , Dipeptides/metabolism , Dose-Response Relationship, Drug , Heterocyclic Compounds, 1-Ring/metabolism , Humans , Lutetium/metabolism , Male , Prostate/metabolism , Prostate-Specific Antigen , Radioisotopes/metabolism , Receptors, Androgen/biosynthesis , Up-Regulation
13.
Respir Res ; 20(1): 106, 2019 May 29.
Article in English | MEDLINE | ID: mdl-31142317

ABSTRACT

BACKGROUND: Tumor chemo-resistance is a hallmark of malignant tumors as well as the major cause of poor survival rates in lung cancer. Transmembrane-4 L-six family member-1 (TM4SF1), an antigen that serves as an oncogene, mainly affects tumor invasion and metastasis. We investigated the roles of TM4SF1 in non-small-cell lung cancer progression, particularly in the regulation of chemo-sensitivity. METHODS: TM4SF1 was silenced by small interfering RNA transfection.TM4SF1 expression in cell lines and tissues were determined by Quantitative Real-time PCR. MTS, clonogenic, Transwell assay, Flow cytometry verified cell function. By RT-PCR, Western blot, the mechanisms were studied. RESULTS: TM4SF1 was upregulated in both lung cancer cell lines and tissues, compared with 293 T epithelial cells. Analysis of online databases revealed that high expression of TM4SF1 is associated with the older patient age, smoking habits, and poor patient survival and outcome. Knockdown of TM4SF1 substantially inhibited tumor cell growth, migration, and invasion, and enhanced the chemo-sensitivity of the lung cancer cell lines A549 and H1299 to cisplatin and paclitaxel. Furthermore, the silencing of TM4SF1 induced lung cancer cell apoptosis and arrested cells at the G2/M phase. These results suggest that TM4SF1 is associated with lung cancer progression and appears to be required for tumor cell growth, maintenance of chemo-resistance and metastasis. We further found that TM4SF1 exerts these effects in part by regulating the expression of the discoidin domain receptor DDR1 and its downstream target, the Akt/ERK/mTOR pathway, and consequently alters cell sensitivity to chemo-reagents and contributes to invasion and metastasis. CONCLUSIONS: These findings demonstrate that TM4SF1 may serve as a prognostic factor for lung cancer chemo-response and patient outcome.


Subject(s)
Antigens, Surface/biosynthesis , Carcinoma, Non-Small-Cell Lung/metabolism , Discoidin Domain Receptor 1/metabolism , Lung Neoplasms/metabolism , MAP Kinase Signaling System/physiology , Neoplasm Proteins/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , A549 Cells , Adult , Aged , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/physiology , Drug Resistance, Neoplasm/physiology , Female , HEK293 Cells , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness/pathology
14.
Can J Cardiol ; 35(2): 208-216, 2019 02.
Article in English | MEDLINE | ID: mdl-30760428

ABSTRACT

BACKGROUND: Myeloid differentiation protein 1 (MD1) is expressed in various tissues, including the heart. However, the role of MD1 in obesity-related atrial remodelling remains incompletely understood. Here, this study intends to determine the regulatory role and underlying mechanisms of MD1 in obesity-related atrial remodelling. METHODS: A high-fat diet (HFD) feeding was performed in 6-week-old MD1-knockout (MD1-KO) mice and wild-type (WT) littermates for 20 weeks. Morphological, biochemical, functional, histological, and electrophysiological studies were conducted at the age of 26 weeks. RESULTS: Our results revealed that the MD1 expression levels were downregulated in the atrium of the HFD-fed induced obesity mice. An increase in body weight, glucose intolerance, hyperlipidemia, and adverse atrial remodelling, such as atrial inflammation and fibrosis, were induced by HFD feeding in WT mice. Vulnerability to atrial fibrillation (AF) was also significantly increased by HFD feeding in WT mice. In addition, these adverse effects caused by HFD-fed induced obesity were further exaggerated in MD1-KO mice compared with WT mice. Mechanistically, MD1-KO activated TLR4/NF-κB signaling pathways, which led to atrial remodelling in mice fed by HFD by increasing the phosphorylation of p65 and IκBα. CONCLUSIONS: Our data suggested that MD1 deficiency played an important role in accelerating the development of inflammatory atrial fibrosis and increasing vulnerability to AF in mice with HFD-fed induced obesity, providing an essential target for improving HFD-induced atrial remodelling.


Subject(s)
Atrial Function, Left/physiology , Atrial Remodeling/physiology , Cardiomyopathies/physiopathology , Gene Expression Regulation , Heart Atria/physiopathology , Membrane Glycoproteins/deficiency , Animals , Antigens, Surface/biosynthesis , Antigens, Surface/genetics , Cardiomyopathies/diagnosis , Cardiomyopathies/genetics , Diet, High-Fat , Disease Models, Animal , Echocardiography , Fibrosis/diagnosis , Fibrosis/genetics , Fibrosis/metabolism , Heart Atria/diagnostic imaging , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Mice, Knockout , RNA/genetics
15.
Pathol Res Pract ; 215(3): 490-498, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30612778

ABSTRACT

BACKGROUND: MFG-E8(Milk fat globule-EGF factor 8), a secreted glycoprotein, plays an exceptional role in various diseases. MFG-E8 overexpression is found in a variety of cancers. However, it remains unclear whether MFG-E8 overexpression is associated with the clinicopathological characteristics and prognosis of human breast cancer. MATERIALS AND METHODS: In this study, we detected the expression and localization of MFG-E8 protein in breast cancer and cancer-adjacent tissues using immunohistochemical staining, Western blot analysis and immunofluorescence. We analyzed the association between MFG-E8 expression and clinical characteristics and outcomes of breast cancer patients with different HR and HER2 statuses. RESULTS: Our results confirmed that MFG-E8 expression increased significantly in breast cancer compared with cancer-adjacent tissues by immunohistochemical staining (P < 0.001). Similarly, the Western blot results further confirmed the increased expression of MFG-E8 in breast cancer compared with cancer-adjacent tissues (P = 0.001). Immunofluorescence staining showed that MFG-E8 was mainly localized in the cytoplasm and membrane of tumor cells, consistent with the immunohistochemical staining results. The high expression levels of MFG-E8 showed a greater association with lymph node metastasis, TNM stage and histological grade (P < 0.001). Moreover, high MFG-E8 expression was related to a shortened overall survival (OS) (P < 0.001) and disease-free survival (DFS) (P < 0.001). Bioinformatics analysis with a Kaplan-Meier plotter also demonstrated a strong association of MFG-E8 mRNA overexpression with a short OS and DFS compared with low MFG-E8 expression (P = 0.040, P = 0.005). CONCLUSIONS: Our findings indicate that MFG-E8 may be a potential marker for poor prognosis and survival in breast cancer.


Subject(s)
Antigens, Surface/biosynthesis , Biomarkers, Tumor/analysis , Breast Neoplasms/pathology , Carcinoma/pathology , Milk Proteins/biosynthesis , Adult , Aged , Breast Neoplasms/mortality , Carcinoma/mortality , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Prognosis
16.
Curr Stem Cell Res Ther ; 14(1): 9-13, 2019.
Article in English | MEDLINE | ID: mdl-30152289

ABSTRACT

Cardiovascular Disease (CVD) is one of the world-wide healthcare problem that involves the heart or blood vessels. CVD includes myocardial infarction and coronary artery diseases (CAD). Dysfunctional myocardial cells are leading causes of low cardiac output or ventricular dysfunction after cardiac arrest and may contribute to the progression of CVD which could not generate new cardiomyocytes in human adult heart. The mesenchymal stem cells (MSCs) which are present in adult marrow can self-renew and have the capacity of differentiation into multiple types of cells including cardiomyocytes. Recent biochemical analyses greatly revealed that several regulators of MSCs, such as HGF, PDGF, Wnt, and Notch-1 signaling pathways have been shown to be involved in the proliferation and differentiation into cardiomyocytes. Preclinical studies are paving the way for further applications of MSCs in the repair of myocardial infarction. In this study, we discuss and summarize the paracrine mechanisms involved in MSCs differentiation into cardiomyocytes.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells/physiology , Myocytes, Cardiac/physiology , Paracrine Communication/physiology , Antigens, Surface/biosynthesis , Cell Proliferation , Cell-Derived Microparticles/physiology , Coronary Artery Disease/therapy , Cytokines/physiology , Exosomes/physiology , Humans , Intercellular Signaling Peptides and Proteins/physiology , Mesenchymal Stem Cell Transplantation/psychology , Mesenchymal Stem Cells/cytology , Myocardial Infarction/therapy , Myocytes, Cardiac/cytology , Proto-Oncogene Proteins c-met/physiology , Receptors, Platelet-Derived Growth Factor/physiology , Wnt Signaling Pathway/physiology
17.
Curr Opin Support Palliat Care ; 12(3): 359-365, 2018 09.
Article in English | MEDLINE | ID: mdl-29939893

ABSTRACT

PURPOSE OF REVIEW: In recent years, major advances in molecular imaging of prostate cancers (PCa) were made with the development and clinical validation of highly accurate PET tracers to stage and restage the disease. Prostate-specific membrane antigen (PSMA) is a transmembrane protein highly expressed in PCa, and its expression has led to the development of PSMA-binding radiopharmaceuticals for molecular imaging or radioligand therapy (RLT). We herein review the recent literature published on diagnostic and therapeutic (i.e. theranostic) PSMA tracers. RECENT FINDINGS: Development in small PSMA-targeted molecules labeled with gallium-68 and fluorine-18 show promising results for primary staging and detection of disease at biochemical recurrence using PET/computed tomography (PET/CT). Studies show a higher sensitivity and specificity, along with an improved detection rate over conventional imaging (CT scan and bone scan) or choline PET tracers, especially for restaging after prostate-specific antigen failure following loco-regional therapy. In addition, some PSMA tracers can be labeled with beta-minus and alpha particle emitters, yielding encouraging response rates and low toxicity, and potentially offering a new line of targeted therapy for metastatic castration-resistant PCa. SUMMARY: PSMA-targeted tracers have shown unprecedented accuracy to stage and restage PCa using PET/CT. Given their specific biodistribution toward PCa tissue, PSMA RLT now offers new therapeutic possibilities to target metastatic PCa. Prospective multicenter randomized studies investigating the clinical impact of PSMA-targeted molecules are urgently needed.


Subject(s)
Antigens, Surface/biosynthesis , Glutamate Carboxypeptidase II/biosynthesis , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/radiotherapy , Fluorine Radioisotopes/administration & dosage , Gallium Radioisotopes/administration & dosage , Humans , Male , Neoplasm Recurrence, Local , Neoplasm Staging , Positron Emission Tomography Computed Tomography , Positron-Emission Tomography , Prospective Studies , Prostatic Neoplasms/pathology , Radiopharmaceuticals/administration & dosage , Theranostic Nanomedicine
18.
Eur Radiol ; 28(12): 5275-5283, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29846803

ABSTRACT

OBJECTIVES: To quantitatively characterize clinically significant intra-prostatic cancer (IPC) by prostate-specific membrane antigen (PSMA) expression and cell density on PSMA-11 positron emission tomography/magnetic resonance (PET/MR). METHODS: Retrospective study approved by the institutional review board with informed written consent obtained. Patients with a solitary, biopsy-proven prostate cancer, Gleason score (GS) ≥7, presenting for initial evaluation by PET/computerised tomography (PET/CT), underwent early prostate PET/MR immediately after PSMA-11 tracer injection. PET/MR [MRI-based attenuation correction (MRAC)] and PET/CT [CT-based AC (CTAC)] maximal standardised uptake value (SUVmax) and minimal and mean apparent diffusion coefficient (ADCmin, ADCmean; respectively) in normal prostatic tissue (NPT) were compared to IPC area. The relationship between SUVmax, ADCmin and ADCmean measurements was obtained. RESULTS: Twenty-two patients (mean age 69.5±5.0 years) were included in the analysis. Forty-four prostate areas were evaluated (22 IPC and 22 NPT). Median MRAC SUVmax of NPT was significantly lower than median MRAC SUVmax of IPC (p < 0.0001). Median ADCmin and ADCmean of NPT was significantly higher than median ADCmin and ADCmean of IPC (p < 0.0001). A very good correlation was found between MRAC SUVmax with CTAC SUVmax (rho = -0.843, p < 0.0001). A good inverse relationship was found between MRAC SUVmax and CTAC SUVmax with ADCmin (rho = -0.717, p < 0.0001 and -0.740, p < 0.0001; respectively; Z = 0.22, p = 0.82, NS) and with MRAC SUVmax and ADCmean (rho = -0.737, p < 0.0001). CONCLUSIONS: PET/MR SUVmax, ADCmin and ADCmean are distinct biomarkers able to differentiate between IPC and NPT in naïve prostate cancer patients with GS ≥ 7. KEY POINTS: • PSMA PET/MR metrics differentiate between normal and tumoural prostatic tissue. • A multi-parametric approach combining molecular and anatomical information might direct prostate biopsy. • PSMA PET/MR metrics are warranted for radiomics analysis.


Subject(s)
Antigens, Surface/biosynthesis , Glutamate Carboxypeptidase II/biosynthesis , Magnetic Resonance Imaging/methods , Positron Emission Tomography Computed Tomography/methods , Prostate/diagnostic imaging , Prostatic Neoplasms/diagnosis , Aged , Biomarkers, Tumor/biosynthesis , Biopsy , Cell Count , Humans , Male , Middle Aged , Neoplasm Grading , Prognosis , Prostate/metabolism , Prostatic Neoplasms/metabolism , Retrospective Studies
19.
Prostate ; 78(10): 758-765, 2018 07.
Article in English | MEDLINE | ID: mdl-29633296

ABSTRACT

BACKGROUND: Prostate-specific membrane antigen (PSMA)-based imaging and therapy are increasingly used for prostate cancer management. However, limitations are a low PSMA expression in certain patients. Androgen receptor axis inhibition can induce PSMA expression in vitro. We hypothesized that different approved compounds upregulate PSMA expression and tested their effect in vitro. METHODS: Androgen receptor (AR) expressing prostate cancer (LNCaP) and epithelial prostate cells (PNT1A) were treated for 7 days with enzalutamide, dutasteride, rapamycin, metformin, lovastatin, and acetylsalicylic acid (ASA). PSMA and AR protein expression was assessed using flow cytometry, immunocytochemistry and immunoblotting. Furthermore, uptake and internalization of 177 Lu-PSMA-617 was performed. RESULTS: Enzalutamide and dutasteride led to a significant (both P < 0.05) upregulation of PSMA surface levels in LNCaP cells. In addition, treatment with rapamycin showed a non-significant trend toward PSMA upregulation. No changes were detected after treatment with vehicle, metformin, lovastatin, and ASA. Total PSMA protein expression was significantly enhanced after treatment with enzalutamide and rapamycin (both P < 0.05), whereas dutasteride led to a non-significant upregulation. Uptake of 177 Lu-PSMA-617 was significantly increased after treatment of LNCaP with enzalutamide, dutasteride, and rapamycin (P < 0.05). In addition, internalization was significantly increased by enzalutamide and rapamycin (P < 0.05), and non-significantly increased by dutasteride. CONCLUSION: In conclusion, our data provide new insights into the effect of different approved pharmacological compounds that can markedly upregulate PSMA expression and radioligand uptake in vitro. Pharmacologically induced PSMA expression may prove useful to improve prostate cancer detection and to enhance anticancer effects in PSMA-based therapy.


Subject(s)
Antigens, Surface/biosynthesis , Antineoplastic Agents/pharmacology , Glutamate Carboxypeptidase II/biosynthesis , Prostate/drug effects , Prostatic Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Male , Prostate/metabolism , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/drug therapy , Up-Regulation/drug effects
20.
Br J Cancer ; 118(6): 813-819, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29462126

ABSTRACT

Background:The intratumoural heterogeneity, often driven by epithelial-to-mesenchymal transition (EMT), significantly contributes to chemoresistance and disease progression in adenocarcinomas. Methods:We introduced a high-throughput screening platform to identify surface antigens that associate with epithelial­mesenchymal plasticity in well-defined pairs of epithelial cell lines and their mesenchymal counterparts. Using multicolour flow cytometry, we then analysed the expression of 10 most robustly changed antigens and identified a 10-molecule surface signature, in pan-cytokeratin-positive/EpCAM-positive and -negative fractions of dissociated breast tumours. Results:We found that surface CD9, CD29, CD49c, and integrin ß5 are lost in breast cancer cells that underwent EMT in vivo. The tetraspanin family member CD9 was concordantly downregulated both in vitro and in vivo and associated with epithelial phenotype and favourable prognosis. Conclusions:We propose that overall landscape of 10-molecule surface signature expression reflects the epithelial­mesenchymal plasticity in breast cancer.


Subject(s)
Antigens, Neoplasm/biosynthesis , Antigens, Surface/biosynthesis , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Antigens, Neoplasm/immunology , Antigens, Surface/immunology , Biomarkers, Tumor , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Plasticity/immunology , Cellular Reprogramming/physiology , Epithelial-Mesenchymal Transition/immunology , Female , Flow Cytometry , High-Throughput Screening Assays , Humans , Neoplasm Metastasis , Tetraspanin 29/biosynthesis , Tetraspanin 29/immunology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...