Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Lab Invest ; 104(3): 100304, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38092179

RESUMO

Gene expression profiling from formalin-fixed paraffin-embedded (FFPE) renal allograft biopsies is a promising approach for feasibly providing a molecular diagnosis of rejection. However, large-scale studies evaluating the performance of models using NanoString platform data to define molecular archetypes of rejection are lacking. We tested a diverse retrospective cohort of over 1400 FFPE biopsy specimens, rescored according to Banff 2019 criteria and representing 10 of 11 United Network of Organ Sharing regions, using the Banff Human Organ Transplant panel from NanoString and developed a multiclass model from the gene expression data to assign relative probabilities of 4 molecular archetypes: No Rejection, Antibody-Mediated Rejection, T Cell-Mediated Rejection, and Mixed Rejection. Using Least Absolute Shrinkage and Selection Operator regularized regression with 10-fold cross-validation fitted to 1050 biopsies in the discovery cohort and technically validated on an additional 345 biopsies, our model achieved overall accuracy of 85% in the discovery cohort and 80% in the validation cohort, with ≥75% positive predictive value for each class, except for the Mixed Rejection class in the validation cohort (positive predictive value, 53%). This study represents the technical validation of the first model built from a large and diverse sample of diagnostic FFPE biopsy specimens to define and classify molecular archetypes of histologically defined diagnoses as derived from Banff Human Organ Transplant panel gene expression profiling data.


Assuntos
Nefropatias , Transplante de Rim , Transplante de Órgãos , Humanos , Transplante de Rim/efeitos adversos , Estudos de Coortes , Estudos Retrospectivos , Rejeição de Enxerto/diagnóstico , Rejeição de Enxerto/genética , Nefropatias/patologia , Expressão Gênica , Biópsia , Rim/patologia
2.
Kidney Int ; 100(6): 1303-1315, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34352311

RESUMO

Kidney failure is common in patients with Coronavirus Disease-19 (COVID-19), resulting in increased morbidity and mortality. In an international collaboration, 284 kidney biopsies were evaluated to improve understanding of kidney disease in COVID-19. Diagnoses were compared to five years of 63,575 native biopsies prior to the pandemic and 13,955 allograft biopsies to identify diseases that have increased in patients with COVID-19. Genotyping for APOL1 G1 and G2 alleles was performed in 107 African American and Hispanic patients. Immunohistochemistry for SARS-CoV-2 was utilized to assess direct viral infection in 273 cases along with clinical information at the time of biopsy. The leading indication for native biopsy was acute kidney injury (45.4%), followed by proteinuria with or without concurrent acute kidney injury (42.6%). There were more African American patients (44.6%) than patients of other ethnicities. The most common diagnosis in native biopsies was collapsing glomerulopathy (25.8%), which was associated with high-risk APOL1 genotypes in 91.7% of cases. Compared to the five-year biopsy database, the frequency of myoglobin cast nephropathy and proliferative glomerulonephritis with monoclonal IgG deposits was also increased in patients with COVID-19 (3.3% and 1.7%, respectively), while there was a reduced frequency of chronic conditions (including diabetes mellitus, IgA nephropathy, and arterionephrosclerosis) as the primary diagnosis. In transplants, the leading indication was acute kidney injury (86.4%), for which rejection was the predominant diagnosis (61.4%). Direct SARS-CoV-2 viral infection was not identified. Thus, our multi-center large case series identified kidney diseases that disproportionately affect patients with COVID-19 and demonstrated a high frequency of APOL1 high-risk genotypes within this group, with no evidence of direct viral infection within the kidney.


Assuntos
Injúria Renal Aguda , COVID-19 , Apolipoproteína L1/genética , Humanos , Rim , Estudos Retrospectivos , SARS-CoV-2
3.
Clin J Am Soc Nephrol ; 16(12): 1790-1796, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-36630401

RESUMO

BACKGROUND AND OBJECTIVES: Black Americans have a higher incidence of kidney disease compared with populations that do not have recent African ancestry. Two risk variants in the APOL1 are responsible for a portion of this higher risk. We sought to assess the odds of AKI conferred by APOL1 risk alleles in patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: Black Americans who tested positive for coronavirus disease 2019 (COVID-19) were genotyped to determine APOL1 risk allele status. We assessed the incidence of AKI, persistent AKI, and AKI requiring KRT within 21 days of the PCR-based diagnosis. Outcomes were adjusted for age, sex, body mass index, hypertension, eGFR, and use of angiotensin-converting enzyme inhibitor/angiotensin receptor blocker. RESULTS: In total, 126 cases of SARS-CoV-2 infection were included within a 5-month period, with 16 (13%) and 110 (87%) cases with two and zero/one APOL1 high-risk alleles, respectively. AKI occurred in 11 (69%) patients with two APOL1 high-risk alleles and 39 (35%) patients with zero/one high-risk alleles (adjusted odds ratio, 4.41; 95% confidence interval, 1.11 to 17.52; P=0.04). Persistent AKI occurred in eight (50%) patients with two APOL1 high-risk alleles and 21 (19%) of those with zero/one high-risk alleles (adjusted odds ratio, 3.53; 95% confidence interval, 1.8 to 11.57; P=0.04). AKI KRT occurred in four (25%) of those with two APOL1 high-risk alleles and eight (7%) of those with zero/one high-risk alleles (adjusted odds ratio, 4.99; 95% confidence interval, 1.02 to 24.4, P=0.05). CONCLUSIONS: APOL1 high-risk alleles are associated with greater odds of AKI in Black American patients with COVID-19.


Assuntos
Injúria Renal Aguda , COVID-19 , Humanos , Negro ou Afro-Americano/genética , Apolipoproteína L1/genética , COVID-19/genética , SARS-CoV-2 , Genótipo , Injúria Renal Aguda/genética , Fatores de Risco , Apolipoproteínas/genética
4.
Kidney360 ; 2(11): 1770-1780, 2021 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-35372991

RESUMO

Background: Immune responses to vaccination are a known trigger for a new onset of glomerular disease or disease flare in susceptible individuals. Mass immunization against SARS-CoV-2 in the COVID-19 pandemic provides a unique opportunity to study vaccination-associated autoimmune kidney diseases. In the recent literature, there are several patient reports demonstrating a temporal association of SARS-CoV-2 immunization and kidney diseases. Methods: Here, we present a series of 29 cases of biopsy-proven glomerular disease in patients recently vaccinated against SARS-CoV-2 and identified patients who developed a new onset of IgA nephropathy, minimal change disease, membranous nephropathy, ANCA-associated GN, collapsing glomerulopathy, or diffuse lupus nephritis diagnosed on kidney biopsies postimmunization, as well as recurrent ANCA-associated GN. This included 28 cases of de novo GN within native kidney biopsies and one disease flare in an allograft. Results: The patients with collapsing glomerulopathy were of Black descent and had two APOL1 genomic risk alleles. A brief literature review of patient reports and small series is also provided to include all reported cases to date (n=52). The incidence of induction of glomerular disease in response to SARS-CoV-2 immunization is unknown; however, there was no overall increase in incidence of glomerular disease when compared with the 2 years prior to the COVID-19 pandemic diagnosed on kidney biopsies in our practice. Conclusions: Glomerular disease to vaccination is rare, although it should be monitored as a potential adverse event.


Assuntos
COVID-19 , Glomerulonefrite por IGA , Apolipoproteína L1 , Vacinas contra COVID-19/efeitos adversos , Glomerulonefrite por IGA/epidemiologia , Humanos , Pandemias , SARS-CoV-2 , Vacinação/efeitos adversos
5.
Cytokine ; 114: 18-25, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30580156

RESUMO

Meprin metalloendopeptidases, comprising α and ß isoforms, are widely expressed in mammalian cells and organs including kidney, intestines, lungs, skin, and bladder, and in a variety of immune cells and cancer cells. Meprins proteolytically process many inflammatory mediators, including cytokines, chemokines, and other bioactive proteins and peptides that control the function of immune cells. The knowledge of meprin-mediated processing of inflammatory mediators and other target substrates provides a pathophysiologic link for the involvement of meprins in the pathogenesis of many inflammatory disorders. Meprins are now known to play important roles in inflammatory diseases including acute kidney injury, sepsis, urinary tract infections, bladder inflammation, and inflammatory bowel disease. The proteolysis of epithelial and endothelial barriers including cell junctional proteins by meprins promotes leukocyte influx into areas of tissue damage to result in inflammation. Meprins degrade extracellular matrix proteins; this ability of meprins is implicated in the cell migration of leukocytes and the invasion of tumor cells that express meprins. Proteolytic processing and maturation of procollagens provides evidence that meprins are involved in collagen maturation and deposition in the fibrotic processes involved in the formation of keloids and hypertrophic scars and lung fibrosis. This review highlights recent progress in understanding the role of meprins in inflammatory disorders in both human and mouse models.


Assuntos
Inflamação/metabolismo , Inflamação/patologia , Metaloproteases/metabolismo , Sequência de Aminoácidos , Animais , Citocinas/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Humanos , Metaloproteases/química , Proteólise
6.
Biol Chem ; 397(11): 1135-1146, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27279059

RESUMO

Kallikrein-related peptidase 7 (KLK7) is a serine protease encoded within the kallikrein gene cluster located on human chromosome region 19q13.3-13.4. KLK7 is overexpressed in human pancreatic ductal adenocarcinomas (PDACs), but not in normal pancreas. Examination of KLK7 mRNA levels in pancreatic cancer cell lines revealed that it is readily detected in MIA PaCa-2 and PK-1 cells, but not in Panc-1 cells. Treatment of Panc-1 cells with the histone deacetylase (HDAC) inhibitor trichostatin A (TSA) significantly induced KLK7 mRNA expression. Similarly, KLK7 is highly expressed in cervical cancer cells, but its expression in the human cervical cancer cell line HeLa is only detected following TSA treatment. Promoter deletion analysis revealed that the proximal -238 promoter region, containing a putative Sp1-binding site, was sufficient for TSA activation of luciferase reporter activity, which was abrogated by the disruption of the Sp1-binding sequence. Consistent with the notion that TSA induced KLK7 expression via Sp1, co-expression of Sp1 with the KLK7-promoter/luciferase construct produced a significant increase in reporter activity. Chromatin immunoprecipitation (ChIP) analysis revealed enriched Sp1 occupancy on the KLK7 promoter following TSA treatment. Similarly, ChIP analysis showed the histone active mark, H3K4Me3, in the KLK7 promoter region was significantly increased after exposure to TSA.


Assuntos
Epigênese Genética , Regulação Neoplásica da Expressão Gênica/genética , Calicreínas/genética , Neoplasias Pancreáticas/patologia , Neoplasias do Colo do Útero/patologia , Linhagem Celular Tumoral , Epigênese Genética/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Neoplasias Pancreáticas/genética , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição Sp1/genética , Transcrição Gênica , Neoplasias do Colo do Útero/genética
7.
Biochem Biophys Rep ; 8: 146-150, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28955950

RESUMO

Monocyte chemotactic protein 1 (CCL2/MCP-1) is a small chemokine involved in the recruitment and trafficking of mononuclear immune cells to inflammation sites. Our studies demonstrate that the metalloendopeptidases meprin A (purified from kidney cortex), recombinant meprin α, and recombinant meprin ß can all process CCL2/MCP-1. The cleavage sites were determined by amino acid sequencing and mass spectrometry analysis of the generated products, and the biological activity of the products was evaluated by chemotactic migration assay using THP-1 cells. The cleavage sites generated by the meprin isoforms revealed that meprin A and meprin α cleaved the N-terminal domain of mouse CCL2/MCP-1 at the Asn6 and Ala7 bond, resulting in significant reduction in the chemotactic activity of the cleaved CCL2/MCP-1. Meprin ß was unable to cleave the N-terminus of mouse CCL2/MCP-1 but cleaved the C-terminal region between Ser74 and Glu75. Human CCL2/MCP-1 that lacks the murine C-terminal region was also cleaved by meprin α at the N-terminus resulting in significant loss of CCL2/MCP-1 biological activity, whereas meprin ß did not affect the biological activity. These studies suggest that meprin α and meprin ß may play important roles in regulating the CCL2/MCP-1 chemokine activity during inflammation.

8.
J Proteomics Bioinform ; 8(7): 149-154, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26430350

RESUMO

BACKGROUND: Whole-pelvis radiation therapy is common practice in the post-surgical treatment of cervical and endometrial cancer. Gastrointestinal mucositis is an adverse side effect of radiation therapy, and is a primary concern in patient management. We investigate whether proteomic information obtained from blood samples drawn from patients scheduled to receive radiation therapy for gynecological cancers could be used to predict which patients are most susceptible to radiation-induced gastrointestinal mucositis, in order to improve the individualization of radiation therapy. METHODS: We use 132 proteins measured on 17 gynecological cancer patients in a convex-hull-based, selective-voting ensemble classifier to classify each patient into one of two classes: patients who would not (class 1) or would (class 2) develop gastrointestinal mucositis. We employ 20 repetitions of 10-fold cross-validation to measure classification accuracy. RESULTS: We achieved a 95% confidence interval on average prediction accuracy of (0.711, 0.771) using pre-radiation proteomic profiles to predict which patients would experience gastrointestinal mucositis. Pathway analysis of the 12 most prominent proteins indicated that they could be assembled into a single interaction network with direct associations. The function associated with the highest number of these 12 proteins was cell-to-cell signaling and interaction. CONCLUSIONS: Pre-radiation proteomic profiles have the potential to classify cervical/endometrial cancer patients with high accuracy as to their susceptibility to gastrointestinal mucositis following radiation therapy. Further study of the network of 12 identified proteins is warranted with a larger patient sample to confirm that these proteins are predictive of gastrointestinal mucositis in this patient population.

9.
PLoS One ; 10(10): e0140025, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26444017

RESUMO

We examined whether endoplasmic reticulum (ER) stress-induced autophagy provides cytoprotection from renal tubular epithelial cell injury due to oxidants and chemical hypoxia in vitro, as well as from ischemia-reperfusion (IR) injury in vivo. We demonstrate that the ER stress inducer tunicamycin triggers an unfolded protein response, upregulates ER chaperone Grp78, and activates the autophagy pathway in renal tubular epithelial cells in culture. Inhibition of ER stress-induced autophagy accelerated caspase-3 activation and cell death suggesting a pro-survival role of ER stress-induced autophagy. Compared to wild-type cells, autophagy-deficient MEFs subjected to ER stress had enhanced caspase-3 activation and cell death, a finding that further supports the cytoprotective role of ER stress-induced autophagy. Induction of autophagy by ER stress markedly afforded cytoprotection from oxidants H2O2 and tert-Butyl hydroperoxide and from chemical hypoxia induced by antimycin A. In contrast, inhibition of ER stress-induced autophagy or autophagy-deficient cells markedly enhanced cell death in response to oxidant injury and chemical hypoxia. In mouse kidney, similarly to renal epithelial cells in culture, tunicamycin triggered ER stress, markedly upregulated Grp78, and activated autophagy without impairing the autophagic flux. In addition, ER stress-induced autophagy markedly ameliorated renal IR injury as evident from significant improvement in renal function and histology. Inhibition of autophagy by chloroquine markedly increased renal IR injury. These studies highlight beneficial impact of ER stress-induced autophagy in renal ischemia-reperfusion injury both in vitro and in vivo.


Assuntos
Autofagia/efeitos dos fármacos , Hipóxia/tratamento farmacológico , Rim/efeitos dos fármacos , Substâncias Protetoras/uso terapêutico , Insuficiência Renal/tratamento farmacológico , Traumatismo por Reperfusão/tratamento farmacológico , Tunicamicina/uso terapêutico , Trifosfato de Adenosina/metabolismo , Animais , Caspases/metabolismo , Linhagem Celular , Citoproteção/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Proteínas de Choque Térmico/metabolismo , Hipóxia/complicações , Hipóxia/metabolismo , Hipóxia/patologia , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxidantes/efeitos adversos , Estresse Oxidativo/efeitos dos fármacos , Insuficiência Renal/complicações , Insuficiência Renal/metabolismo , Insuficiência Renal/patologia , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Resposta a Proteínas não Dobradas/efeitos dos fármacos
10.
Cancer Biol Ther ; 16(10): 1557-65, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26176765

RESUMO

To develop new diagnostic and therapeutic tools to specifically target pancreatic tumors, it is necessary to identify cell-surface proteins that may serve as potential tumor-specific targets. In this study we used an azido-labeled bioorthogonal chemical reporter to metabolically label N-linked glycoproteins on the surface of pancreatic cancer cell lines to identify potential targets that may be exploited for detection and/or treatment of pancreatic cancer. Labeled glycoproteins were tagged with biotin using click chemistry, purified by streptavidin-coupled magnetic beads, separated by gel electrophoresis, and identified by liquid chromatography-tandem mass spectrometry (MS). MS/MS analysis of peptides from 3 cell lines revealed 954 unique proteins enriched in the azido sugar samples relative to control sugar samples. A comparison of the proteins identified in each sample indicated 20% of these proteins were present in 2 cell lines (193 of 954) and 17 of the proteins were found in all 3 cell lines. Five of the 17 proteins identified in all 3 cell lines have not been previously reported to be expressed in pancreatic cancer; thus indicating that novel cell-surface proteins can be revealed through glycoprotein profiling. Western analysis of one of these glycoproteins, ecto-5'-nucleotidase (NT5E), revealed it is expressed in 8 out of 8 pancreatic cancer cell lines examined. Further, immunohistochemical analysis of human pancreatic tissues indicates NT5E is significantly overexpressed in pancreatic tumors compared to normal pancreas. Thus, we have demonstrated that metabolic labeling with bioorthogonal chemical reporters can be used to selectively enrich and identify novel cell-surface glycoproteins expressed in pancreatic ductal adenocarcinomas.


Assuntos
Proteínas de Membrana/metabolismo , Neoplasias Pancreáticas/imunologia , Biomarcadores Tumorais/metabolismo , Humanos , Neoplasias Pancreáticas/patologia
11.
Toxicol Lett ; 236(2): 110-6, 2015 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-25957482

RESUMO

Meprins are oligomeric metalloproteinases that are abundantly expressed in the brush-border membranes of renal proximal tubules. During acute kidney injury (AKI) induced by cisplatin or ischemia-reperfusion, membrane-bound meprins are shed and their localization is altered from the apical membranes toward the basolateral surface of the proximal tubules. Meprins are capable of cleaving basement membrane proteins in vitro, however, it is not known whether meprins are able to degrade extracellular matrix proteins under pathophysiological conditions in vivo. The present study demonstrates that a basement membrane protein, nidogen-1, is cleaved and excreted in the urine of mice subjected to cisplatin-induced nephrotoxicity, a model of AKI. Cleaved nidogen-1 was not detected in the urine of untreated mice, but during the progression of cisplatin nephrotoxicity, the excretion of cleaved nidogen-1 increased in a time-dependent manner. The meprin inhibitor actinonin markedly prevented urinary excretion of the cleaved nidogen-1. In addition, meprin ß-deficient mice, but not meprin α-deficient mice, subjected to cisplatin nephrotoxicity significantly suppressed excretion of cleaved nidogen-1, further suggesting that meprin ß is involved in the cleavage of nidogen-1. These studies provide strong evidence for a pathophysiological link between meprin ß and urinary excretion of cleaved nidogen-1 during cisplatin-induced AKI.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Membrana Basal/metabolismo , Cisplatino/toxicidade , Glicoproteínas de Membrana/metabolismo , Metaloendopeptidases/metabolismo , Injúria Renal Aguda/metabolismo , Animais , Antineoplásicos/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Genótipo , Ácidos Hidroxâmicos , Masculino , Metaloendopeptidases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transporte Proteico
12.
Cancer Biol Ther ; 16(5): 714-23, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25996841

RESUMO

Increased aerobic glycolysis and de novo lipid biosynthesis are common characteristics of invasive cancers. UDP-glucuronosyltransferases (UGTs) are phase II drug metabolizing enzymes that in normal cells possess the ability to glucuronidate these lipids and speed their excretion; however, de-regulation of these enzymes in cancer cells can lead to an accumulation of bioactive lipids, which further fuels cancer progression. We hypothesize that UGT2B isoform expression is down-regulated in cancer cells and that exogenous re-introduction of these enzymes will reduce lipid content, change the cellular phenotype, and inhibit cancer cell proliferation. In this study, steady-state mRNA levels of UGT isoforms from the 2B family were measured using qPCR in 4 breast cancer and 5 pancreatic cancer cell lines. Expression plasmids for UGT2B isoforms known to glucuronidate cellular lipids, UGT2B4, 2B7, and 2B15 were transfected into MCF-7 and Panc-1 cells, and the cytotoxic effects of these enzymes were analyzed using trypan blue exclusion, annexin V/PI staining, TUNEL assays, and caspase-3 immunohistochemistry. There was a significant decrease in cell proliferation and a significant increase in the number of dead cells after transfection with each of the 3 UGT isoforms in both cell lines. Cellular lipids were also found to be significantly decreased after transfection. The results presented here support our hypothesis and emphasize the important role UGTs can play in cellular proliferation and lipid homeostasis. Evaluating the effect of UGT expression on the lipid levels in cancer cell lines can be relevant to understanding the complex regulation of cancer cells, identifying the roles of UGTs as "lipid-controllers" in cellular homeostasis, and illustrating their suitability as targets for future clinical therapy development.


Assuntos
Neoplasias da Mama/genética , Glucuronosiltransferase/genética , Neoplasias Pancreáticas/genética , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Células MCF-7 , Transfecção
13.
J Biol Chem ; 289(19): 13308-22, 2014 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-24662289

RESUMO

Meprin A, composed of α and ß subunits, is a membrane-bound metalloproteinase in renal proximal tubules. Meprin A plays an important role in tubular epithelial cell injury during acute kidney injury (AKI). The present study demonstrated that during ischemia-reperfusion-induced AKI, meprin A was shed from proximal tubule membranes, as evident from its redistribution toward the basolateral side, proteolytic processing in the membranes, and excretion in the urine. To identify the proteolytic enzyme responsible for shedding of meprin A, we generated stable HEK cell lines expressing meprin ß alone and both meprin α and meprin ß for the expression of meprin A. Phorbol 12-myristate 13-acetate and ionomycin stimulated ectodomain shedding of meprin ß and meprin A. Among the inhibitors of various proteases, the broad spectrum inhibitor of the ADAM family of proteases, tumor necrosis factor-α protease inhibitor (TAPI-1), was most effective in preventing constitutive, phorbol 12-myristate 13-acetate-, and ionomycin-stimulated shedding of meprin ß and meprin A in the medium of both transfectants. The use of differential inhibitors for ADAM10 and ADAM17 indicated that ADAM10 inhibition is sufficient to block shedding. In agreement with these results, small interfering RNA to ADAM10 but not to ADAM9 or ADAM17 inhibited meprin ß and meprin A shedding. Furthermore, overexpression of ADAM10 resulted in enhanced shedding of meprin ß from both transfectants. Our studies demonstrate that ADAM10 is the major ADAM metalloproteinase responsible for the constitutive and stimulated shedding of meprin ß and meprin A. These studies further suggest that inhibiting ADAM 10 activity could be of therapeutic benefit in AKI.


Assuntos
Proteínas ADAM/metabolismo , Injúria Renal Aguda/enzimologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Membrana Celular/enzimologia , Proteínas de Membrana/metabolismo , Metaloendopeptidases/metabolismo , Proteínas ADAM/genética , Proteína ADAM10 , Proteína ADAM17 , Injúria Renal Aguda/genética , Injúria Renal Aguda/patologia , Secretases da Proteína Precursora do Amiloide/genética , Animais , Ionóforos de Cálcio/farmacologia , Carcinógenos/farmacologia , Membrana Celular/genética , Membrana Celular/patologia , Células HEK293 , Humanos , Ionomicina/farmacologia , Masculino , Proteínas de Membrana/genética , Metaloendopeptidases/genética , Camundongos , Acetato de Tetradecanoilforbol/farmacologia
14.
Methods Mol Biol ; 1133: 141-54, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24567100

RESUMO

Members of the caspase family of proteases are evolutionarily conserved cysteine proteases that play a crucial role as the central executioners of the apoptotic pathway. Since the discovery of caspases, many methods have been developed to detect their activation and are widely used in basic and clinical studies. In a mouse tissue, caspase activation can be monitored by cleavage of caspase-specific synthetic substrates and by detecting cleaved caspase by western blot analysis of the tissue extract. In tissue sections, active caspase can be detected by immunostaining using specific antibodies to the active caspase. In addition, among the myriads of caspase-specific substrates known so far, cleaved fragments produced by caspases from the substrates such as PARP, lamin A, and cytokeratin-18 can be monitored in tissue sections by immunostaining as well as western blots of tissue extracts. In general, more than one method should be used to ascertain detection of activation of caspases in a mouse tissue.


Assuntos
Apoptose/genética , Caspases/isolamento & purificação , Biologia Molecular/métodos , Animais , Caspases/genética , Camundongos
15.
J Proteomics Bioinform ; Suppl 10: S10003, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25635161

RESUMO

BACKGROUND: The development of novel targeted cancer therapies and/or diagnostic tools is dependent upon an understanding of the differential expression of molecular targets between normal tissues and tumors. Many of these potential targets are cell-surface receptors; however, our knowledge of the cell-surface proteins upregulated in pancreatic tumors is limited, thus impeding the development of targeted therapies for pancreatic cancer. To develop new diagnostic and therapeutic tools to specifically target pancreatic tumors, we sought to identify cell-surface proteins that may serve as potential tumor-specfic targets. METHODS: Membrane glycoproteins on the pancreatic cancer cell lines BxPC-3 were labeled with the bifunctional linker biocytin hydrazide. Following proteolytic digestion, biotinylated glycopeptides were captured with streptavidin-coupled beads then released by PNGaseF-mediated endoglycosidase cleavage and identified by liquid chromatography-tandem mass spectrometry (MS). A protein identified by the cell-surface glycoprotein capture procedure, CD109, was evaluated by western analysis of lysates of pancreatic cancer cell lines and by immunohistochemistry in sections of pancreatic ductal adenocarcinoma and non- neoplastic pancreatic tissues. RESULTS: MS/MS analysis of glycopeptides captured from BxPC-3 cells revealed 18 proteins predicted or known to be associated with the plasma membrane, including CD109, which has not been reported in pancreatic cancer. Western analysis of CD109 in lysates prepared from pancreatic cancer cell lines revealed it was expressed in 6 of 8 cell lines, with a high level of expression in BxPC-3, MIAPaCa-2, and Panc-1 cells. Immunohistochemical analyses of human pancreatic tissues indicate CD109 is significantly overexpressed in pancreatic tumors compared to normal pancreas. CONCLUSIONS: The selective capture of glycopeptides from the surface of pancreatic cancer cell lines can reveal novel cell-surface glycoproteins expressed in pancreatic ductal adenocarcinomas.

16.
PLoS One ; 8(8): e72580, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24013211

RESUMO

BACKGROUND: Serotonin (5-HT) is a biogenic amine that also acts as a mitogen and a developmental signal early in rodent embryogenesis. Genetic and pharmacological disruption of 5-HT signaling causes various diseases and disorders via mediating central nervous system, cardiovascular system, and serious abnormalities on a growing embryo. Today, neither the effective modulators on 5-HT signaling pathways nor the genes affected by 5-HT signal are well known yet. METHODOLOGY/PRINCIPAL FINDINGS: In an attempt to identify the genes altered by 5-HT signaling pathways, we analyzed the global gene expression via the Illumina array platform using the mouse WG-6 v2.0 Expression BeadChip containing 45,281 probe sets representing 30,854 genes in megakaryocytes isolated from mice infused with 5-HT or saline. We identified 723 differentially expressed genes of which 706 were induced and 17 were repressed by elevated plasma 5-HT. CONCLUSIONS/SIGNIFICANCE: Hierarchical gene clustering analysis was utilized to represent relations between groups and clusters. Using gene ontology mining tools and canonical pathway analyses, we identified multiple biological pathways that are regulated by 5-HT: (i) cytoskeletal remodeling, (ii) G-protein signaling, (iii) vesicular transport, and (iv) apoptosis and survival. Our data encompass the first extensive genome-wide based profiling in the progenitors of platelets in response to 5-HT elevation in vivo.


Assuntos
Regulação da Expressão Gênica/fisiologia , Megacariócitos/metabolismo , Serotonina/sangue , Animais , Perfilação da Expressão Gênica/métodos , Masculino , Megacariócitos/citologia , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos
17.
BMC Cancer ; 13: 404, 2013 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-24007603

RESUMO

BACKGROUND: The identification of new serum biomarkers with high sensitivity and specificity is an important priority in pancreatic cancer research. Through an extensive proteomics analysis of pancreatic cancer cell lines and pancreatic juice, we previously generated a list of candidate pancreatic cancer biomarkers. The present study details further validation of four of our previously identified candidates: regenerating islet-derived 1 beta (REG1B), syncollin (SYCN), anterior gradient homolog 2 protein (AGR2), and lysyl oxidase-like 2 (LOXL2). METHODS: The candidate biomarkers were validated using enzyme-linked immunosorbent assays in two sample sets of serum/plasma comprising a total of 432 samples (Sample Set A: pancreatic ductal adenocarcinoma (PDAC, n = 100), healthy (n = 92); Sample Set B: PDAC (n = 82), benign (n = 41), disease-free (n = 47), other cancers (n = 70)). Biomarker performance in distinguishing PDAC from each control group was assessed individually in the two sample sets. Subsequently, multiparametric modeling was applied to assess the ability of all possible two and three marker panels in distinguishing PDAC from disease-free controls. The models were generated using sample set B, and then validated in Sample Set A. RESULTS: Individually, all markers were significantly elevated in PDAC compared to healthy controls in at least one sample set (p ≤ 0.01). SYCN, REG1B and AGR2 were also significantly elevated in PDAC compared to benign controls (p ≤ 0.01), and AGR2 was significantly elevated in PDAC compared to other cancers (p < 0.01). CA19.9 was also assessed. Individually, CA19.9 showed the greatest area under the curve (AUC) in receiver operating characteristic (ROC) analysis when compared to the tested candidates; however when analyzed in combination, three panels (CA19.9 + REG1B (AUC of 0.88), CA19.9 + SYCN + REG1B (AUC of 0.87) and CA19.9 + AGR2 + REG1B (AUC of 0.87)) showed an AUC that was significantly greater (p < 0.05) than that of CA19.9 alone (AUC of 0.82). In a comparison of early-stage (Stage I-II) PDAC to disease free controls, the combination of SYCN + REG1B + CA19.9 showed the greatest AUC in both sample sets, (AUC of 0.87 and 0.92 in Sets A and B, respectively). CONCLUSIONS: Additional serum biomarkers, particularly SYCN and REG1B, when combined with CA19.9, show promise as improved diagnostic indicators of pancreatic cancer, which therefore warrants further validation.


Assuntos
Biomarcadores Tumorais/sangue , Antígeno CA-19-9/sangue , Proteínas de Transporte/sangue , Litostatina/sangue , Proteínas de Membrana/sangue , Neoplasias Pancreáticas/sangue , Neoplasias Pancreáticas/diagnóstico , Adulto , Fatores Etários , Idoso , Aminoácido Oxirredutases/sangue , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mucoproteínas , Estadiamento de Neoplasias , Proteínas Oncogênicas , Proteínas/metabolismo , Curva ROC , Reprodutibilidade dos Testes , Estudos Retrospectivos , Fatores Sexuais
18.
Am J Physiol Renal Physiol ; 304(9): F1150-8, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23427141

RESUMO

Meprin A, composed of α- and ß-subunits, is a membrane-associated neutral metalloendoprotease that belongs to the astacin family of zinc endopeptidases. It was first discovered as an azocasein and benzoyl-l-tyrosyl-p-aminobenzoic acid hydrolase in the brush-border membranes of proximal tubules and intestines. Meprin isoforms are now found to be widely distributed in various organs (kidney, intestines, leukocytes, skin, bladder, and a variety of cancer cells) and are capable of hydrolyzing and processing a large number of substrates, including extracellular matrix proteins, cytokines, adherens junction proteins, hormones, bioactive peptides, and cell surface proteins. The ability of meprin A to cleave various substrates sheds new light on the functional properties of this enzyme, including matrix remodeling, inflammation, and cell-cell and cell-matrix processes. Following ischemia-reperfusion (IR)- and cisplatin-induced acute kidney injury (AKI), meprin A is redistributed toward the basolateral plasma membrane, and the cleaved form of meprin A is excreted in the urine. These studies suggest that altered localization and shedding of meprin A in places other than the apical membranes may be deleterious in vivo in acute tubular injury. These studies also provide new insight into the importance of a sheddase involved in the release of membrane-associated meprin A under pathological conditions. Meprin A is injurious to the kidney during AKI, as meprin A-knockout mice and meprin inhibition provide protective roles and improve renal function. Meprin A, therefore, plays an important role in AKI and potentially is a unique target for therapeutic intervention during AKI.


Assuntos
Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Metaloendopeptidases/fisiologia , Animais , Membrana Celular/patologia , Membrana Celular/fisiologia , Modelos Animais de Doenças , Células Epiteliais/patologia , Células Epiteliais/fisiologia , Humanos , Leucócitos/patologia , Leucócitos/fisiologia , Metaloendopeptidases/genética , Camundongos , Camundongos Knockout , Ratos
19.
Cancer Epidemiol Biomarkers Prev ; 21(7): 1135-42, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22573795

RESUMO

BACKGROUND: Kallikrein-related peptidase 7 (KLK7) is overexpressed in pancreatic ductal adenocarcinomas (PDAC). The aims of this study were to examine the expression of KLK7 during progression of pancreatic intraepithelial neoplasia (PanIN) to invasive PDAC and to assess its prognostic significance for PDAC. METHODS: Immunohistochemistry was used to assess KLK7 expression using a tissue microarray (TMA) and full sections of pancreatic tissue containing normal tissue, PanIN, and invasive adenocarcinoma, and the association between KLK7 expression and prognosis was examined by a population-based pancreatic cancer TMA. RESULTS: Normal pancreatic epithelium was negative for KLK7 in either TMAs or full sections. Analysis by TMAs showed that 91% of cases showed KLK7 positivity in the adenocarcinoma component, which was significantly higher than PanIN 2/3. In full tissue sections of PDAC, KLK7 expression was detected in less than 1% of cells among PanIN 1A lesions, and increased with grade among PanIN 1B and PanIN2/3 lesions before reaching 69% in the invasive PDAC. In patients with unresected PDAC, KLK7 positivity was significantly associated with shorter overall survival. CONCLUSIONS: Aberrant KLK7 expression starts in intermediate-to-late stages of PanIN progression, and KLK7-positive staining is associated with almost a three-fold increase in mortality rate of patients with unresected PDAC. IMPACT: The association of KLK7 expression and poor outcome of patients with unresectable PDAC suggests that inhibiting either KLK7 expression and/or activity could be a therapeutic strategy. Because the vast majority of patients present with unresectable disease, such an intervention could have a significant impact upon the overall survival of this patient population.


Assuntos
Adenocarcinoma/mortalidade , Biomarcadores Tumorais/metabolismo , Carcinoma in Situ/mortalidade , Carcinoma Ductal Pancreático/mortalidade , Calicreínas/metabolismo , Neoplasias Pancreáticas/mortalidade , Lesões Pré-Cancerosas/mortalidade , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Idoso , Carcinoma in Situ/metabolismo , Carcinoma in Situ/patologia , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Progressão da Doença , Feminino , Humanos , Técnicas Imunoenzimáticas , Masculino , Pessoa de Meia-Idade , Pâncreas/metabolismo , Pâncreas/patologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Prognóstico , Taxa de Sobrevida , Análise Serial de Tecidos
20.
Exp Brain Res ; 212(1): 109-17, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21626096

RESUMO

The malfunction of tight junctions (TJs) between endothelial cells in the blood brain barrier (BBB) is the pathophysiological basis for cerebral ischemia-reperfusion (IR) injury. Claudins, major molecular elements of the TJs, play a key role in the paracellular permeability of the BBB. Although several studies have demonstrated the impact of hyperbaric oxygenation (HBO) on boosting oxygen supply and reducing infarct size, its effect and underlying mechanism on the integrity of the BBB is unknown. To study the function of HBO on claudins and the permeability of the BBB, we replicated the animal model of local cerebral IR. Using Evans blue dye, permeability of the BBB was examined. Transmission electron microscopy (TEM), immunohistochemistry, western blot, and gelatin zymography were used to detect the integrity of the BBB, the expression of claudin-1 and claudin-5, and the activity of matrix metalloproteinases (MMPs) in brain microvessel endothelium. Our data indicate that compared with the sham-operated group, IR increased permeability of the BBB to Evans blue dye (P < 0.01), peaking at 4 h. The BBB ultrastructure was disrupted and the expression of claudin-5 and claudin-1 decreased (P < 0.01) in the 4 and 72 h IR group, respectively. Increased claudin-5 and claudin-1 expression and decreased permeability of the BBB were observed in the HBO + IR group (P < 0.01) via the suppression of MMP-2 and MMP-9, respectively. Our study provides direct evidence that HBO decreases the permeability of the BBB by reducing the enzymatic activity of MMPs and augmenting the expression of claudins at different stages in cerebral IR injury.


Assuntos
Isquemia Encefálica/metabolismo , Claudinas/biossíntese , Oxigenoterapia Hiperbárica , Proteínas de Membrana/biossíntese , Traumatismo por Reperfusão/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Isquemia Encefálica/genética , Isquemia Encefálica/terapia , Claudina-1 , Claudina-5 , Claudinas/genética , Regulação da Expressão Gênica , Oxigenoterapia Hiperbárica/métodos , Masculino , Proteínas de Membrana/genética , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/terapia , Junções Íntimas/genética , Junções Íntimas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA