Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Mol Cell Proteomics ; 23(3): 100722, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38272115

ABSTRACT

Glioblastoma (GBM) is the most aggressive brain tumor and different efforts have been employed in the search for new drugs and therapeutic protocols for GBM. Epitranscriptomics has shed light on new druggable Epigenetic therapies specifically designed to modulate GBM biology and behavior such as Histone Deacetylase inhibitors (iHDAC). Although the effects of iHDAC on GBM have been largely explored, there is a lack of information on the underlaying mechanisms HDAC-dependent that modulate the repertoire of GBM secreted molecules focusing on the set of Extracellular Matrix (ECM) associated proteins, the Matrisome, that may impact the surrounding tumor microenvironment. To acquire a better comprehension of the impacts of HDAC activity on the GBM Matrisome, we studied the alterations on the Matrisome-associated ECM regulators, Core Matrisome ECM glycoproteins, ECM-affiliated proteins and Proteoglycans upon HDAC inhibition in vitro as well as their relationship with glioma pathophysiological/clinical features and angiogenesis. For this, U87MG GBM cells were treated for with iHDAC or vehicle (control) and the whole secretome was processed by Mass Spectrometry NANOLC-MS/MS. In silico analyses revealed that proteins associated to the Angiogenic Matrisome (AngioMatrix), including Decorin, ADAM10, ADAM12 and ADAM15 were differentially regulated in iHDAC versus control secretome. Interestingly, genes coding for the Matrisome proteins differentially regulated were found mutated in patients and were correlated to glioma pathophysiological/clinical features. In vitro functional assays, using HBMEC endothelial cells exposed to the secretome of control or iHDAC treated GBM cells, coupled to 2D and 3D GBM cell culture system, showed impaired migratory capacity of endothelial cells and disrupted tubulogenesis in a Fibronectin and VEGF independent fashion. Collectively, our study provides understanding of epigenetic mechanisms HDAC-dependent to key Matrisomal proteins that may contribute to identify new druggable Epigenetic therapies or gliomagenesis biomarkers with relevant implications to improve therapeutic protocols for this malignancy.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Humans , Glioblastoma/genetics , Glioblastoma/pathology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Endothelial Cells/metabolism , Tandem Mass Spectrometry , Extracellular Matrix/metabolism , Glioma/metabolism , Epigenesis, Genetic , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Brain Neoplasms/drug therapy , Tumor Microenvironment , Membrane Proteins/metabolism , ADAM Proteins/metabolism
2.
Genes (Basel) ; 14(6)2023 05 29.
Article in English | MEDLINE | ID: mdl-37372361

ABSTRACT

Many reproductive physiological processes, such as folliculogenesis, ovulation, implantation, and fertilization, require the synthesis, remodeling, and degradation of the extracellular matrix (ECM). The ADAMTS (A Disintegrin and Metalloproteinase with Thrombospondin Motifs) family genes code for key metalloproteinases in the remodeling process of different ECM. Several genes of this family encode for proteins with important functions in reproductive processes; in particular, ADAMTS1, 4, 5 and 9 are genes that are differentially expressed in cell types and the physiological stages of reproductive tissues. ADAMTS enzymes degrade proteoglycans in the ECM of the follicles so that the oocytes can be released and regulate follicle development during folliculogenesis, favoring the action of essential growth factors, such as FGF-2, FGF-7 and GDF-9. The transcriptional regulation of ADAMTS1 and 9 in preovulatory follicles occurs because of the gonadotropin surge in preovulatory follicles, via the progesterone/progesterone receptor complex. In addition, in the case of ADAMTS1, pathways involving protein kinase A (PKA), extracellular signal regulated protein kinase (ERK1/2) and the epidermal growth factor receptor (EGFR) might contribute to ECM regulation. Different Omic studies indicate the importance of genes of the ADAMTS family from a reproductive aspect. ADAMTS genes could serve as biomarkers for genetic improvement and contribute to enhance fertility and animal reproduction; however, more research related to these genes, the synthesis of proteins encoded by these genes, and regulation in farm animals is needed.


Subject(s)
ADAM Proteins , ADAMTS Proteins , Female , Animals , ADAMTS Proteins/genetics , ADAM Proteins/genetics , ADAM Proteins/metabolism , Ovulation/genetics , Oocytes/metabolism , Progesterone
3.
Clin Transl Oncol ; 25(5): 1218-1241, 2023 May.
Article in English | MEDLINE | ID: mdl-36897508

ABSTRACT

Breast cancer (BC) is one of the most prevalent types of cancer in women. Despite advancement in early detection and efficient treatment, recurrence and metastasis continue to pose a significant risk to the life of BC patients. Brain metastasis (BM) reported in 17-20 percent of BC patients is considered as a major cause of mortality and morbidity in these patients. BM includes various steps from primary breast tumor to secondary tumor formation. Various steps involved are primary tumor formation, angiogenesis, invasion, extravasation, and brain colonization. Genes involved in different pathways have been reported to be associated with BC cells metastasizing to the brain. ADAM8 gene, EN1 transcription factor, WNT, and VEGF signaling pathway have been associated with primary breast tumor; MMP1, COX2, XCR4, PI3k/Akt, ERK and MAPK pathways in angiogenesis; Noth, CD44, Zo-1, CEMIP, S0X2 and OLIG2 are involved in invasion, extravasation and colonization, respectively. In addition, the blood-brain barrier is also a key factor in BM. Dysregulation of cell junctions, tumor microenvironment and loss of function of microglia leads to BBB disruption ultimately resulting in BM. Various therapeutic strategies are currently used to control the BM in BC. Oncolytic virus therapy, immune checkpoint inhibitors, mTOR-PI3k inhibitors and immunotherapy have been developed to target various genes involved in BM in BC. In addition, RNA interference (RNAi) and CRISPR/Cas9 are novel interventions in the field of BCBM where research to validate these and clinical trials are being carried out. Gaining a better knowledge of metastasis biology is critical for establishing better treatment methods and attaining long-term therapeutic efficacies against BC. The current review has been compiled with an aim to evaluate the role of various genes and signaling pathways involved in multiple steps of BM in BC. The therapeutic strategies being used currently and the novel ones being explored to control BM in BC have also been discussed at length.


Subject(s)
Brain Neoplasms , Breast Neoplasms , Humans , Female , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Breast Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Brain Neoplasms/metabolism , Signal Transduction/genetics , Tumor Microenvironment , Membrane Proteins/metabolism , ADAM Proteins/metabolism
4.
Clin Transl Oncol ; 25(2): 473-481, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36183312

ABSTRACT

BACKGROUND: ADAM metallopeptidase domain 12 (ADAM12) is generally upregulated in tissues of various tumors, emerging as a prognostic biomarker. However, the clinical significance of serum ADAM12 in tumors still remains to be fully elucidated. The present study aimed to investigate the expression and prognostic value of serum ADAM12 in tumor patients. MATERIALS AND METHODS: Serum samples were collected from healthy doners (HDs; n = 87) and patients (n = 238) with a clinical diagnosis of breast, liver, lung, stomach and esophageal (STES) and thyroid cancer. Serum ADAM12 protein and mRNA expression was detected by enzyme-linked immunosorbent assay (ELISA) and reverse transcription polymerase chain reaction (RT-PCR), respectively. Receiver-operator characteristic (ROC) analysis was performed to explored the prognostic value of serum ADAM12 expression. RESULTS: The expression of serum ADAM12 in breast and liver cancer patients was significantly upregulated compared with HDs. In patients with breast cancer, the levels of serum ADAM12 protein and mRNA were significantly higher in tumor stages than that in HDs (p < 0.05), with AUC value of 0.82. In liver cancer, elevated levels of serum ADAM12 protein were significantly correlated with clinical stage (r = 0.74; p = 6.9e-4) and T stage (r = 0.74, p = 7.6e-4), and attained AUC value of 1. However, the clinical significance of serum ADAM12 expression in lung, STES and thyroid cancer had not been found. CONCLUSIONS: Serum ADAM12 expression showed high degree of tumor heterogeneity, and may be a valuable noninvasive diagnostic and prognostic biomarker for breast and liver cancer.


Subject(s)
Breast Neoplasms , Liver Neoplasms , Thyroid Neoplasms , Humans , Female , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM12 Protein , Breast Neoplasms/genetics , Liver Neoplasms/diagnosis , RNA, Messenger/metabolism , Biomarkers
5.
Exp Cell Res ; 398(2): 112415, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33296662

ABSTRACT

A Disintegrin And Metalloprotease 23 (ADAM23) is a member of the ADAMs family of transmembrane proteins, mostly expressed in nervous system, and involved in traffic and stabilization of Kv1-potassium channels, synaptic transmission, neurite outgrowth, neuronal morphology and cell adhesion. Also, ADAM23 has been linked to human pathological conditions, such as epilepsy, cancer metastasis and cardiomyopathy. ADAM23 functionality depends on the molecule presence at the cell surface and along the secretory pathway, as expected for a cell surface receptor. Because endocytosis is an important functional regulatory mechanism of plasma membrane receptors and no information is available about the traffic or turnover of non-catalytic ADAMs, we investigated ADAM23 internalization, recycling and half-life properties. Here, we show that ADAM23 undergoes constitutive internalization from the plasma membrane, a process that depends on lipid raft integrity, and is redistributed to intracellular vesicles, especially early and recycling endosomes. Furthermore, we observed that ADAM23 is recycled from intracellular compartments back to the plasma membrane and thus has longer half-life and higher cell surface stability compared with other ADAMs. Our findings suggest that regulation of ADAM23 endocytosis/stability could be exploited therapeutically in diseases in which ADAM23 is directly involved, such as epilepsy, cancer progression and cardiac hypertrophy.


Subject(s)
ADAM Proteins/metabolism , Endocytosis , Cell Membrane/metabolism , Cells, Cultured , Endosomes/metabolism , Half-Life , Humans , Membrane Microdomains/metabolism
6.
J Cell Biochem ; 119(10): 8204-8219, 2018 11.
Article in English | MEDLINE | ID: mdl-29923217

ABSTRACT

Osteoblast differentiation is controlled by transcription factor RUNX2 which temporally activates or represses several bone-related genes, including those encoding extracellular matrix proteins or factors that control cell-cell, and cell-matrix interactions. Cell-cell communication in the many skeletal pericellular micro-niches is critical for bone development and involves paracrine secretion of growth factors and morphogens. This paracrine signaling is in part regulated by "A Disintegrin And Metalloproteinase" (ADAM) proteins. These cell membrane-associated metalloproteinases support proteolytic release ("shedding") of protein ectodomains residing at the cell surface. We analyzed microarray and RNA-sequencing data for Adam genes and show that Adam17, Adam10, and Adam9 are stimulated during BMP2 mediated induction of osteogenic differentiation and are robustly expressed in human osteoblastic cells. ADAM17, which was initially identified as a tumor necrosis factor alpha (TNFα) converting enzyme also called (TACE), regulates TNFα-signaling pathway, which inhibits osteoblast differentiation. We demonstrate that Adam17 expression is suppressed by RUNX2 during osteoblast differentiation through the proximal Adam17 promoter region (-0.4 kb) containing two functional RUNX2 binding motifs. Adam17 downregulation during osteoblast differentiation is paralleled by increased RUNX2 expression, cytoplasmic-nuclear translocation and enhanced binding to the Adam17 proximal promoter. Forced expression of Adam17 reduces Runx2 and Alpl expression, indicating that Adam17 may negatively modulate osteoblast differentiation. These findings suggest a novel regulatory mechanism involving a reciprocal Runx2-Adam17 negative feedback loop to regulate progression through osteoblast differentiation. Our results suggest that RUNX2 may control paracrine signaling through regulation of ectodomain shedding at the cell surface of osteoblasts by directly suppressing Adam17 expression.


Subject(s)
ADAM17 Protein/genetics , Bone Morphogenetic Protein 2/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Feedback, Physiological , Osteoblasts/metabolism , Osteogenesis/genetics , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , ADAM17 Protein/metabolism , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Binding Sites , Bone Morphogenetic Protein 2/metabolism , Cell Differentiation , Cell Line , Cell Line, Tumor , Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Profiling , Gene Expression Regulation , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Osteoblasts/cytology , Paracrine Communication/genetics , Promoter Regions, Genetic , Protein Binding , Rats , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
7.
Neuroscience ; 384: 165-177, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29792904

ABSTRACT

A disintegrin and metalloprotease protein 23 (ADAM23) is a transmembrane type I glycoprotein involved with the development and maintenance of the nervous system, including neurite outgrowth, neuronal adhesion and differentiation and regulation of synaptic transmission. In addition, ADAM23 seems to participate in immune response and tumor establishment through interaction with different members of integrin receptors. Here, we describe a novel monoclonal antibody (DL11C8) that specifically recognizes the cysteine-rich domain of both pre-protein (100 kDa) and mature (70 kDa) forms of ADAM23 from different species, including human, rodents and avian orthologs. Using this antibody, we detected both forms of ADAM23 on the cell surface of three neuronal cell lineages (Neuro-2a, SH-SY5Y and CHLA-20), with a higher relative content of ADAM23100 kDa. Furthermore, we demonstrate for the first time that a catalytically inactive member of the ADAM family is present in the membrane signaling platforms, namely lipid rafts. Indeed, the mature ADAM2370 kDa partitions between raft and non-raft membrane domains, while the pro-protein ADAM23100 kDa is mainly expressed in non-raft domains. These membranous distributions were observed in both different brain regions homogenates and primary cultured neurons lysates from mouse cortex and cerebellum. Taken together, these findings point out ADAM23 as a lipid raft molecular component.


Subject(s)
ADAM Proteins/metabolism , Membrane Microdomains/metabolism , Animals , Antibodies, Monoclonal , Cell Line, Tumor , Humans , Mice
8.
Biol Reprod ; 99(3): 590-599, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29659700

ABSTRACT

The extracellular matrix (ECM) is a group of molecules that offer structural and biochemical support to cells and interact with them to regulate their function. Also, growth factors (GFs) stored in the ECM can be locally released during ECM remodeling. Here, we hypothesize that the balance between ECM components and remodelers is regulated according to the ovarian steroid milieu to which the oviduct is exposed during the periovulatory period. Follicular growth was manipulated to generate cows that ovulated small follicles (SF-small corpus luteum [SCL]; n = 20) or large follicles (LF-large corpus luteum [LCL]; n = 21) and possess corresponding Estradiol (E2) and Progesterone (P4) plasmatic concentrations. Ampulla and isthmus samples were collected on day 4 (day 0 = ovulation induction) and immediately frozen or fixed. The transcriptional profile (n = 3/group) was evaluated by RNA sequencing. MMP Antibody Array was used to quantify ECM remodelers' protein abundance and immunohistochemistry to quantify type I collagen. Transcriptome analysis revealed the over-representation of ECM organization and remodeling pathways in the LF-LCL group. Transcription of ECM components (collagens), remodelers (ADAMs and MMPs), and related GFs were upregulated in LF-LCL. Protein intensities for MMP3, MMP8, MMP9, MMP13, and TIMP4 were greater for the LF-LCL group. Type I collagen content in the mucosa was greater in SF-SCL group. In conclusion, that the earlier and more intense exposure to E2 and P4 during the periovulatory period in LF-LCL animals stimulates ECM remodeling. We speculate that differential ECM regulation may contribute to oviductal receptivity to the embryo.


Subject(s)
Extracellular Matrix/physiology , Gonadal Steroid Hormones/physiology , Oviducts/physiology , ADAM Proteins/metabolism , Animals , Cattle , Collagen Type I/biosynthesis , Collagen Type I/genetics , Computational Biology , Estradiol/blood , Extracellular Matrix/ultrastructure , Female , Gene Expression Regulation, Developmental , Immunohistochemistry , Matrix Metalloproteinases/metabolism , Ovarian Follicle/physiology , Ovarian Follicle/ultrastructure , Oviducts/ultrastructure , Ovulation/physiology , Pregnancy , Progesterone/blood
9.
Clin Transl Oncol ; 19(1): 58-66, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27026568

ABSTRACT

BACKGROUND: Clinically useful marker molecules for the progression of gastroesophageal reflux disease and Barrett's esophagus (BE) to esophageal adenocarcinoma (EAC) are lacking. Many adenocarcinomas and inflammatory conditions exhibit increased expression of ADAMs, 'a disintegrin and metalloproteinases'. METHODS: We assessed the expression of five ADAMs (9, 10, 12, 17, 19) in three esophageal cell lines (Het-1A, OE19, OE33) by RT-PCR and Western blotting, and in human samples of normal esophagus, esophagitis, BE, Barrett's dysplasia, and EAC by RT-PCR, and in selected samples by immunohistochemistry. RESULTS: EAC patients showed increased mRNA expression of ADAMs 9, 12, 17 and 19, as compared to controls. At immunohistochemistry, ADAM9 and ADAM10 proteins were increased in EAC. Patient samples also showed increased mRNA expression of ADAM12 in esophagitis, of ADAM9 in BE, and of ADAMs 9, 12 and 19 in Barrett's dysplasia, as compared to controls. Two EAC cell lines showed increased ADAM9 mRNA. CONCLUSIONS: ADAM9 expression is increased in EAC. Its predecessors show increased ADAM9 mRNA expression. The importance of the alterations in ADAM expression for the development of EAC, and their use as marker molecules, warrant further studies.


Subject(s)
ADAM Proteins/metabolism , Adenocarcinoma/metabolism , Barrett Esophagus/metabolism , Biomarkers, Tumor/metabolism , Disintegrins/metabolism , Esophageal Neoplasms/metabolism , Gastroesophageal Reflux/metabolism , ADAM Proteins/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Apoptosis , Barrett Esophagus/genetics , Barrett Esophagus/pathology , Biomarkers, Tumor/genetics , Blotting, Western , Case-Control Studies , Cell Proliferation , Disease Progression , Disintegrins/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Female , Follow-Up Studies , Gastroesophageal Reflux/genetics , Gastroesophageal Reflux/pathology , Humans , Immunoenzyme Techniques , Male , Middle Aged , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
10.
J Ovarian Res ; 9: 9, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26916548

ABSTRACT

BACKGROUND: Ovarian carcinomas, usually associated with sex hormones dysregulation, are the leading cause of gynecological neoplastic death. In normal ovaries, hormones play a central role in regulating cell proliferation, differentiation, and apoptosis. On the other hand, hormonal alterations also play a variety of roles in cancer. Stimulation by sex hormones potentially affects gene expression, invasiveness, cell growth and angiogenesis. Proteases of the "a disintegrin and metalloproteinase with thrombospondin motifs" (ADAMTS) family are secreted by different cell types and become involved in collagen processing, cleavage of the proteoglycan matrix, and angiogenesis. We evaluated whether sex hormones affect ADAMTS 1 and 4 expression in ovarian cancer cells. METHODS: We analysed mRNA and protein levels in human ovarian tumor cells with different degrees of malignancy, NIH-OVCAR-3 and ES-2, that were treated or not with estrogen, testosterone and progesterone. RESULTS: Our results suggest that progesterone increases ADAMTS protein and mRNA levels in the lysates from ES-2 cells, and it increases ADAMTS protein in the lysates and conditioned media from NIH-OVCAR-3. Progesterone effects were reversed by RU486 treatment. CONCLUSION: We conclude that progesterone acts via the progesterone receptor to modulate ADAMTS 1 and 4 levels in ovarian cancer cell lines.


Subject(s)
ADAM Proteins/metabolism , Procollagen N-Endopeptidase/metabolism , Progesterone/physiology , Receptors, Progesterone/metabolism , ADAM Proteins/genetics , ADAMTS1 Protein , ADAMTS4 Protein , Cell Line, Tumor , Enzyme Induction , Female , Gene Expression , Humans , Mifepristone/pharmacology , Ovarian Neoplasms , Procollagen N-Endopeptidase/genetics , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism
11.
Curr Vasc Pharmacol ; 14(1): 106-15, 2016.
Article in English | MEDLINE | ID: mdl-26463982

ABSTRACT

Angiogenesis is a key process by which new capillary blood vessels are formed, sustaining the supply of oxygen and other nutrients to the body allowing its growth and wound healing, among others. However, angiogenesis also associates with pathological processes, such us tumor growth. Vascular endothelial cells produce different matrix remodeling enzymes such as matrix metalloproteinases and a-disintegrin and metalloproteinases, which have both positive and negative effects on angiogenesis, regulating the cell environment and signaling. However, little is known about the regulation of the activity of these proteases during vascular development. Reversion-inducing cysteine-rich protein with Kazal motifs (RECK) is a membrane-anchored inhibitor of different matrix metalloproteinases and a-disintegrin and metalloproteinases, being a critical regulator of extracellular matrix remodeling and signaling pathway, particularly Notch, which is critical for the maturation of the growing vessels. Reck knockout mice die in utero showing vascular developmental defects and massive hemorrhages. These defects were not observed in knockout mice for secreted-soluble matrix metalloproteinase inhibitors pointing to an exclusive role of RECK in vascular development and maturation since its location at the plasma membrane. Despite the above, the exact role of RECK in this process has not been clarified. This review is focused to summarize the available information on the role of RECK as membrane anchored matrix metalloproteinases and a-disintegrin and metalloproteinases inhibitor, proposing a hypothesis by which RECK play key roles in the physiology and pathophysiology of the angiogenesis processes.


Subject(s)
GPI-Linked Proteins/metabolism , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/physiology , ADAM Proteins/metabolism , Animals , Extracellular Matrix/metabolism , Humans , Matrix Metalloproteinases/metabolism , Mice , Mice, Knockout , Signal Transduction/physiology
12.
Bol Asoc Med P R ; 107(2): 86-8, 2015.
Article in English | MEDLINE | ID: mdl-26434092

ABSTRACT

We present a case of a 61 year old man who presented with a 3 week history of easy bruising and ecchymoses in both thighs and right arm without significant trauma. Physical exam was remarkable for oozing gums, diminished second heart sound, a systolic ejection murmur at the aortic position with radiation to carotids, and delayed pulses. Laboratories were remarkable for iron deficiency anemia. Echocardiogram was consistent with severe aortic stenosis. Colonoscopy revealed several arteriovenous malformations throughout the colon. There is an association between severe aortic stenosis and gastrointestinal bleeding. The pathogenesis of Heyde's Syndrome involves iron deficiency anemia due to acquired von Willebrand factor (vWF) deficiency and ultimately gastrointestinal angiodysplasia. Correct diagnosis and management warrants a multidisciplinary approach.


Subject(s)
ADAM Proteins/metabolism , Angiodysplasia/etiology , Aortic Valve Stenosis/complications , Gastrointestinal Hemorrhage/etiology , von Willebrand Diseases/etiology , ADAMTS13 Protein , Angiodysplasia/diagnosis , Aortic Valve Stenosis/physiopathology , Colonoscopy , Diabetic Nephropathies/complications , Ecchymosis/etiology , Enzyme Activation , Humans , Hypertension/complications , Male , Middle Aged , Protein Stability , Protein Structure, Quaternary , Shear Strength , Syndrome , Treatment Refusal , von Willebrand Diseases/diagnosis , von Willebrand Factor/chemistry
13.
Am J Physiol Heart Circ Physiol ; 309(5): H926-34, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26254330

ABSTRACT

We previously reported that type 2 angiotensin-converting enzyme (ACE2) compensatory activity is impaired by the disintegrin and metalloprotease 17 (ADAM17), and lack of ACE2 is associated with oxidative stress in neurogenic hypertension. To investigate the relationship between ADAM17 and oxidative stress, Neuro2A cells were treated with ANG II (100 nM) 24 h after vehicle or α-lipoic acid (LA, 500 µM). ADAM17 expression was increased by ANG II (120.5 ± 9.1 vs. 100.2 ± 0.8%, P < 0.05) and decreased after LA (69.0 ± 0.3 vs. 120.5 ± 9.1%, P < 0.05). In another set of experiments, LA reduced ADAM17 (92.9 ± 5.3 vs. 100.0 ± 11.2%, P < 0.05) following its overexpression. Moreover, ADAM17 activity was reduced by LA in ADAM17-overexpressing cells [109.5 ± 19.8 vs. 158.0 ± 20.0 fluorescence units (FU)·min(-1)·µg protein(-1), P < 0.05], in which ADAM17 overexpression increased oxidative stress (114.1 ± 2.5 vs. 101.0 ± 1.0%, P < 0.05). Conversely, LA-treated cells attenuated ADAM17 overexpression-induced oxidative stress (76.0 ± 9.1 vs. 114.1 ± 2.5%, P < 0.05). In deoxycorticosterone acetate (DOCA)-salt hypertensive mice, a model in which ADAM17 expression and activity are increased, hypertension was blunted by pretreatment with LA (119.0 ± 2.4 vs. 131.4 ± 2.2 mmHg, P < 0.05). In addition, LA improved dysautonomia and baroreflex sensitivity. Furthermore, LA blunted the increase in NADPH oxidase subunit expression, as well as the increase in ADAM17 and decrease in ACE2 activity in the hypothalamus of DOCA-salt hypertensive mice. Taken together, these data suggest that LA might preserve ACE2 compensatory activity by breaking the feedforward cycle between ADAM17 and oxidative stress, resulting in a reduction of neurogenic hypertension.


Subject(s)
ADAM Proteins/metabolism , Antioxidants/pharmacology , Hypertension/metabolism , Oxidative Stress , Thioctic Acid/pharmacology , ADAM Proteins/genetics , ADAM17 Protein , Angiotensin II/pharmacology , Angiotensin-Converting Enzyme 2 , Animals , Antioxidants/therapeutic use , Baroreflex , Cell Line, Tumor , Hypertension/drug therapy , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , NADPH Oxidases/metabolism , Neurons/drug effects , Neurons/metabolism , Peptidyl-Dipeptidase A/metabolism , Thioctic Acid/therapeutic use
14.
PLoS One ; 10(7): e0132784, 2015.
Article in English | MEDLINE | ID: mdl-26168189

ABSTRACT

We have investigated whether von Willebrand factor, ADAMTS13 (a disintegrin and metalloproteinase with thrombospondin type 1 motif, member 13), and D-Dimer were associated with different levels of renal function in patients with type 1 diabetes. Patients were classified according to level of renal function through estimated glomerular filtration rate: ≥90 and <130mL/min/1,73m2, n=52 (control group), ≥60 and <90mL/min/1,73m2, n=29 (mild renal dysfunction group), <60mL/min/1,73m2, n=28 (severe renal dysfunction group); and through urinary albumin excretion: normoalbuminuria, microalbuminuria and macroalbuminuria. Von Willebrand factor, ADAMTS13, and D-Dimer plasma levels were determined by enzyme-linked immunosorbent assay. ADAMTS13 activity was determined by fluorescence resonance energy transfer assay. Von Willebrand factor levels were increased in patients with mild (P=0.001) and severe (P<0.001) renal dysfunction as compared to the control group. ADAMTS13 levels were also increased in mild (P=0.029) and severe (P=0.002) renal dysfunction groups in comparison to the control group, while ADAMTS13 activity was increased only in the severe renal dysfunction group as compared to the control group (P=0.006). No significant differences were observed among the groups regarding von Willebrand factor/ADAMTS13 ratio. ADAMTS13 activity/ADAMTS13 levels ratio was reduced in patients with mild (P=0.013) and severe (P=0.015) renal dysfunction as compared to the control group. D-Dimer levels were increased in patients with mild (P=0.006) and severe (P<0.001) renal dysfunction as compared to the control group; it was also higher in patients with severe renal dysfunction as compared to the mild renal dysfunction group (P=0.019). Similar results were found for albuminuria classification. Increased von Willebrand factor, ADAMTS13, and D-Dimer levels and decreased ADAMTS13 activity/ADAMTS13 levels ratio are associated with renal dysfunction in patients with type 1 diabetes, suggesting that endothelial dysfunction and hypercoagulability are associated with nephropathy in type 1 diabetes.


Subject(s)
ADAM Proteins/metabolism , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/metabolism , Diabetic Nephropathies/metabolism , Fibrin Fibrinogen Degradation Products/metabolism , von Willebrand Factor/metabolism , ADAMTS13 Protein , Adult , Diabetic Nephropathies/complications , Diabetic Nephropathies/pathology , Female , Humans , Male , Middle Aged
15.
Neuroscience ; 304: 340-8, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26197225

ABSTRACT

Oxidative stress is a major risk factor for Alzheimer's disease (AD) that has been suggested to be the trigger of AD pathology. However, whether oxidative damage precedes and contributes directly to the intracellular accumulation of beta amyloid 1-42 (ßA42) peptide remains a matter of debate. Chronic exposure to low doses of ozone similar to the levels during a day of high pollution in México City causes a state of oxidative stress that elicits progressive neurodegeneration in the hippocampi of rats. Several reports have demonstrated that the mitochondria are among the first organelles to be affected by oxidative stress and ßA42 toxicity and act as sites of the accumulation of ßA42, which affects energy metabolism. However, the mechanisms related to the neurodegeneration process and organelle damage that occur in conditions of chronic exposure to low doses of ozone have not been demonstrated. To analyze the effect of chronic ozone chronic exposure on changes in the production and accumulation of the ßA42 and ßA40 peptides in the mitochondria of hippocampal neurons of rats exposed to ozone, we examined the mitochondrial expression levels of Presenilins 1 and 2 and ADAM10 to detect changes related to the oxidative stress caused by low doses of ozone (0.25ppm). The results revealed significant accumulations of ßA42 peptide in the mitochondrial fractions on days 60 and 90 of ozone exposure along with reductions in beta amyloid 1-40 accumulation, significant overexpressions of Pres2 and significant reductions in ADAM10 expression. Beta amyloid immunodetection revealed that there were some intracellular deposits of ßA42 and that ßA42 and the mitochondrial markers OPA1 and COX1 colocalized. These results indicate that the time of exposure to ozone and the accumulation of ßA42 in the mitochondria of the hippocampal cells of rats were correlated. Our results suggest that the accumulation of the ßA42 peptide may promote mitochondrial dysfunction due to its accumulation and overproduction.


Subject(s)
Amyloid beta-Peptides/metabolism , Hippocampus/metabolism , Mitochondria/metabolism , Oxidative Stress/physiology , Ozone/toxicity , Peptide Fragments/metabolism , ADAM Proteins/metabolism , ADAM10 Protein , Air Pollutants , Animals , Chronic Disease , Disease Models, Animal , Hippocampus/pathology , Male , Mitochondria/pathology , Presenilin-1/metabolism , Presenilin-2/metabolism , Random Allocation , Rats, Wistar , Signal Transduction/physiology
16.
Cell Adh Migr ; 9(4): 293-9, 2015.
Article in English | MEDLINE | ID: mdl-26211476

ABSTRACT

One of the most important features of malignant cells is their capacity to invade adjacent tissues and metastasize to distant organs. This process involves the creation, by tumor and stroma cells, of a specific microenvironment, suitable for proliferation, migration and invasion of tumor cells. The ADAM family of proteins has been involved in these processes. This work aimed to investigate the role of the recombinant disintegrin domain of the human ADAM9 (rADAM9D) on the adhesive and mobility properties of DU145 prostate tumor cells. rADAM9D was able to support DU145 cell adhesion, inhibit the migration of DU145 cells, as well as the invasion of this cell line through matrigel in vitro. Overall this work demonstrates that rADAM9D induces specific cellular migratory properties when compared with different constructs having additional domains, specially those of metalloproteinase and cysteine-rich domains. Furthermore, we showed that rADAM9D was able to inhibit cell adhesion, migration and invasion mainly through interacting with α6ß1 in DU145 tumor cell line. These results may contribute to the development of new therapeutic strategies for prostate cancer.


Subject(s)
ADAM Proteins/metabolism , Disintegrins/metabolism , Membrane Proteins/metabolism , Prostatic Neoplasms/metabolism , ADAM Proteins/genetics , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Disintegrins/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Male , Membrane Proteins/genetics , Neoplasm Invasiveness , Prostatic Neoplasms/genetics
17.
Int. arch. otorhinolaryngol. (Impr.) ; 19(2): 112-115, Apr-Jun/2015. tab
Article in English | LILACS | ID: lil-747142

ABSTRACT

Introduction Parotid gland incidentalomas (PGIs) are unexpected hypermetabolic foci in the parotid region that can be found when scanning with whole-body positron emission/computed tomography (PET/CT). These deposits are most commonly due to benign lesions such as Warthin tumor. Objective The aim of this study was to determine the prevalence of PGIs identified in PET/CT scans and to assess the role of smoking in their etiology. Methods We retrospectively reviewed all PET/CT scans performed at our center in search of PGIs and identified smoking status and standardized uptake value (SUVmax) in each case. We also analyzed the database of parotidectomies performed in our department in the previous 10 years and focused on the pathologic diagnosis and the presence or absence of smoking in each case. Results Sixteen cases of PGIs were found in 4,250 PET/CT scans, accounting for 0.4% . The average SUVmax was 6.5 (range 2.8 to 16). Cytology was performed in five patients; it was benign in four cases and inconclusive in one case. Thirteen patients had a history of smoking. Of the parotidectomies performed in our center with a diagnosis of Warthin tumor, we identified a history of smoking in 93.8% of those patients. Conclusions The prevalence of PGIs on PET/CT was similar to that reported by other authors. Warthin tumor is frequently diagnosed among PGIs on PET/CT, and it has a strong relationship with smoking. We suggest that a diagnosis other than Warthin tumor should be considered for PGIs in nonsmokers. .


Subject(s)
Humans , ADAM Proteins/metabolism , Proteolysis , von Willebrand Factor/chemistry , von Willebrand Factor/metabolism , Binding Sites , Calcium/metabolism , Disulfides/chemistry , Disulfides/metabolism , Hydrogen Bonding , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Stability , Protein Structure, Tertiary , Protein Isoforms/chemistry , Protein Isoforms/metabolism , von Willebrand Factor/genetics
18.
Genet Mol Res ; 14(2): 4391-8, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25966212

ABSTRACT

We examined disintegrin and metalloproteinase 17 (ADAM17) protein expression in esophageal squamous cell carcinoma and its clinical and pathological correlated factors. Western blotting and immunohistochemistry were used to detect ADAM17 protein expression in esophageal squamous cell carcinoma and the corresponding normal esophageal mucosa in 50 cases. ADAM17 protein expression in 50 cases with esophageal squamous cells was 0.887 ± 0.174; the positive expression rate was 66% (33/50). ADAM17 protein expression in corresponding normal esophageal mucosa was 0.273 ± 0.081; the positive expression rate was 6% (3/50). Expression in esophageal squamous cell carcinoma was significantly higher than that in the normal esophageal group (P < 0.01). Esophageal squamous cell ADAM17 protein expression and the positive rate were correlated with lymph node metastasis and TNM stage (P < 0.05), but not correlated with gender, age, and histological grade (P > 0.05). ADAM17 protein was highly expressed in esophageal squamous cell carcinoma. This protein may play an important role in the incidence, invasion, and metastasis of esophageal cancer and is valuable for the prognosis of patients with esophageal cancer.


Subject(s)
ADAM Proteins/metabolism , Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , ADAM17 Protein , Aged , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Female , Humans , Lymphatic Metastasis , Male , Middle Aged
19.
Clin Transl Oncol ; 17(8): 604-11, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25786367

ABSTRACT

BACKGROUNDS: A disintegrin and metalloproteinase (ADAM) 17 has been indicated to be an indispensable regulator of cellular events from proliferation to migration. Although prognostic importance of ADAM17 expression has been investigated in several tumours, its clinical utility as a useful prognostic molecular marker remains unclear in gastric cancer. In the current study, we evaluated the expression of ADAM17 and its prognostic significance in gastric cancer patients after curative gastrectomy. METHODS: The prognostic significance of ADAM17 expression was analysed immunohistochemically in 156 patients with gastric cancer who had undergone curative gastrectomy, and the relationship between its expression and clinicopathological factors was also evaluated. RESULTS: High ADAM17 expression was detected in 79 patients (51 %), whereas low expression was found in 77 cases (49 %). There was significant correlation between gender, histology, lymph node metastasis, vascular invasion, the presence of recurrence and high ADAM17 expression. Recurrence in patients with high ADAM17 expression was significantly higher than that for patients with low ADAM17 expression (p = 0.032). The median disease-free survival (DFS) time for patients with tumours with high ADAM17 expression was worse than that of patients with tumours with low ADAM17 expression (16.6 vs. 44.2 months, p = 0.004). In addition, patients with low ADAM17 expression had a higher median overall survival (OS) (49.6 vs. 26.9 months, p = 0.019) compared to those with high ADAM17 expression. Multivariate analysis indicated that the rate of ADAM17 expression was an independent prognostic factor for DFS, in addition to the already known important clinicopathological prognostic indicator. But the prognostic importance of ADAM17 expression could not be proved by multivariate analysis for OS. CONCLUSIONS: The potential value of ADAM17 expression as a useful molecular marker in gastric cancer progression should be evaluated comprehensively; it may predict recurrence and poor prognosis in patients with gastric cancer after curative resection.


Subject(s)
ADAM Proteins/metabolism , Adenocarcinoma, Mucinous/secondary , Adenocarcinoma/secondary , Carcinoma, Signet Ring Cell/secondary , Gastrectomy/mortality , Neoplasm Recurrence, Local/pathology , Stomach Neoplasms/pathology , ADAM17 Protein , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/surgery , Adenocarcinoma, Mucinous/metabolism , Adenocarcinoma, Mucinous/mortality , Adenocarcinoma, Mucinous/surgery , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Carcinoma, Signet Ring Cell/metabolism , Carcinoma, Signet Ring Cell/mortality , Carcinoma, Signet Ring Cell/surgery , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/surgery , Neoplasm Staging , Prognosis , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Stomach Neoplasms/surgery , Survival Rate
20.
Oncogene ; 34(10): 1270-9, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-24662834

ABSTRACT

Intratumoral heterogeneity (ITH) represents an obstacle for cancer diagnosis and treatment, but little is known about its functional role in cancer progression. The A Desintegrin And Metalloproteinase 23 (ADAM23) gene is epigenetically silenced in different types of tumors, and silencing is often associated with advanced disease and metastasis. Here, we show that invasive breast tumors exhibit significant ADAM23-ITH and that this heterogeneity is critical for tumor growth and metastasis. We demonstrate that while loss of ADAM23 expression enhances invasion, it causes a severe proliferative deficiency and is not itself sufficient to trigger metastasis. Rather, we observed that, in ADAM23-heterotypic environments, ADAM23-negative cells promote tumor growth and metastasis by enhancing the proliferation and invasion of adjacent A23-positive cells through the production of LGI4 (Leucine-rich Glioma Inactivated 4) and nitric oxide (NO). Ablation of LGI4 and NO in A23-negative cells significantly attenuates A23-positive cell proliferation and invasion. Our work denotes a driving role of ADAM23-ITH during disease progression, shifting the malignant phenotype from the cellular to the tissue level. Our findings also provide insights for therapeutic intervention, enforcing the need to ascertain ITH to improve cancer diagnosis and therapy.


Subject(s)
ADAM Proteins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Extracellular Matrix Proteins/metabolism , Nitric Oxide/metabolism , ADAM Proteins/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , DNA Methylation , Epigenesis, Genetic , Extracellular Matrix Proteins/genetics , Female , Gene Silencing , Humans , Neoplasm Metastasis , Nerve Tissue Proteins , Tumor Burden , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL