Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Cell Mol Gastroenterol Hepatol ; 11(3): 783-801, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33069918

RESUMO

BACKGROUND & AIMS: Tight junctions form a barrier to the paracellular passage of luminal antigens. Although most tight junction proteins reside within the apical tight junction complex, claudin-18 localizes mainly to the basolateral membrane where its contribution to paracellular ion transport is undefined. Claudin-18 loss in mice results in gastric neoplasia development and tumorigenesis that may or may not be due to tight junction dysfunction. The aim here was to investigate paracellular permeability defects in stomach mucosa from claudin-18 knockout (Cldn18-KO) mice. METHODS: Stomach tissue from wild-type, heterozygous, or Cldn18-KO mice were stripped of the external muscle layer and mounted in Ussing chambers. Transepithelial resistance, dextran 4 kDa flux, and potential difference (PD) were calculated from the chambered tissues after identifying differences in tissue histopathology that were used to normalize these measurements. Marker expression for claudins and ion transporters were investigated by transcriptomic and immunostaining analysis. RESULTS: No paracellular permeability defects were evident in stomach mucosa from Cldn18-KO mice. RNAseq identified changes in 4 claudins from Cldn18-KO mice, particularly the up-regulation of claudin-2. Although claudin-2 localized to tight junctions in cells at the base of gastric glands, its presence did not contribute overall to mucosal permeability. Stomach tissue from Cldn18-KO mice also had no PD versus a lumen-negative PD in tissues from wild-type mice. This difference resulted from changes in transcellular Cl- permeability with the down-regulation of Cl- loading and Cl- secreting anion transporters. CONCLUSIONS: Our findings suggest that Cldn18-KO has no effect on tight junction permeability in the stomach from adult mice but rather affects anion permeability. The phenotype in these mice may thus be secondary to transcellular anion transporter expression/function in the absence of claudin-18.


Assuntos
Cloretos/metabolismo , Claudinas/deficiência , Células Epiteliais/metabolismo , Mucosa Gástrica/metabolismo , Junções Íntimas/metabolismo , Animais , Permeabilidade da Membrana Celular , Claudinas/genética , Claudinas/metabolismo , Células Epiteliais/citologia , Feminino , Mucosa Gástrica/citologia , Íons/metabolismo , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , RNA-Seq , Regulação para Cima
2.
Nucleic Acids Res ; 48(19): 10615-10631, 2020 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-32776089

RESUMO

Lowering of prion protein (PrP) expression in the brain is a genetically validated therapeutic hypothesis in prion disease. We recently showed that antisense oligonucleotide (ASO)-mediated PrP suppression extends survival and delays disease onset in intracerebrally prion-infected mice in both prophylactic and delayed dosing paradigms. Here, we examine the efficacy of this therapeutic approach across diverse paradigms, varying the dose and dosing regimen, prion strain, treatment timepoint, and examining symptomatic, survival, and biomarker readouts. We recapitulate our previous findings with additional PrP-targeting ASOs, and demonstrate therapeutic benefit against four additional prion strains. We demonstrate that <25% PrP suppression is sufficient to extend survival and delay symptoms in a prophylactic paradigm. Rise in both neuroinflammation and neuronal injury markers can be reversed by a single dose of PrP-lowering ASO administered after the detection of pathological change. Chronic ASO-mediated suppression of PrP beginning at any time up to early signs of neuropathology confers benefit similar to constitutive heterozygous PrP knockout. Remarkably, even after emergence of frank symptoms including weight loss, a single treatment prolongs survival by months in a subset of animals. These results support ASO-mediated PrP lowering, and PrP-lowering therapeutics in general, as a promising path forward against prion disease.


Assuntos
Oligonucleotídeos Antissenso/uso terapêutico , Doenças Priônicas/terapia , Proteínas Priônicas/genética , Terapêutica com RNAi/métodos , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular , Camundongos , Camundongos Endogâmicos C57BL , Oligonucleotídeos Antissenso/química , Proteínas Priônicas/metabolismo
3.
Vet Med Sci ; 6(1): 39-43, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31729197

RESUMO

To determine the prevalence of antibodies to Brucella melitensis, Brucella abortus and Coxiella burnetii in animals on Caribbean islands we obtained sera from convenience samples of cattle (C), sheep (S), goats (G) and cats (F) from Dominica (C, S, G), Grenada (C, S, G), Montserrat (C, S, G), Puerto Rico (C), Nevis (C, S, G), St Kitts (C, S, G, F) and St Lucia (C, G). The sera were tested for antibodies against the Brucella spp. using commercial ELISA kits. Some sera were also tested at 1/80 for antibodies to C. burnetii using an indirect fluorescent antibody test. Positive sera were also tested at 1/640. None of 599 cattle, 462 sheep or 434 goats were positive in the Brucella ELISAs. None of 230 cattle had antibodies against C. burnetii, but one of 299 sheep was positive at 1/80 (Dominica - 1/54, 2%, 95% CI (0%-5.6%)), as were two of 314 goats, at 1/80 (Grenada - 1/53, 2%, 95% CI (0%-7.5%)) and 1/640 (St Kitts - 1/18, 5.6%, 95% CI (0%-16.7%)), and one of 34 cats, at 1/80 (St Kitts - 1/34; 3%, 95% CI (0%-8.8%)). Our data suggests that there is a very low prevalence or absence of B. melitensis and B. abortus on Caribbean islands. Coxiella burnetii, however, is present but it appears to be present on only some islands and then only at low levels. Overall, there appears to be a low threat to human and animal health from these organisms in the Caribbean.


Assuntos
Brucelose/veterinária , Doenças do Gato/epidemiologia , Doenças dos Bovinos/epidemiologia , Doenças das Cabras/epidemiologia , Febre Q/veterinária , Doenças dos Ovinos/epidemiologia , Animais , Brucella abortus , Brucella melitensis , Brucelose/epidemiologia , Brucelose/microbiologia , Brucelose Bovina/epidemiologia , Brucelose Bovina/microbiologia , Doenças do Gato/microbiologia , Gatos , Bovinos , Doenças dos Bovinos/microbiologia , Coxiella burnetii , Doenças das Cabras/microbiologia , Cabras , Prevalência , Febre Q/epidemiologia , Febre Q/microbiologia , Estudos Soroepidemiológicos , Ovinos , Doenças dos Ovinos/microbiologia , Carneiro Doméstico , Índias Ocidentais/epidemiologia
4.
JCI Insight ; 52019 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-31361599

RESUMO

Prion disease is a fatal, incurable neurodegenerative disease of humans and other mammals caused by conversion of cellular prion protein (PrP; PrPC) into a self-propagating neurotoxic conformer (prions; PrPSc). Strong genetic proofs of concept support lowering PrP expression as a therapeutic strategy. Antisense oligonucleotides (ASOs) can provide a practical route to lowering one target mRNA in the brain, but their development for prion disease has been hindered by three unresolved questions from prior work: uncertainty about mechanism of action, unclear potential for efficacy against established prion infection, and poor tolerability of drug delivery by osmotic pumps. Here we test antisense oligonucleotides (ASOs) delivered by bolus intracerebroventricular injection to intracerebrally prion-infected wild-type mice. Prophylactic treatments given every 2-3 months extended survival times 61-98%, and a single injection at 120 days post-infection, near the onset of clinical signs, extended survival 55% (87 days). In contrast, a non-targeting control ASO was ineffective. Thus, PrP lowering is the mechanism of action of ASOs effective against prion disease in vivo, and infrequent, or even single, bolus injections of ASOs can slow prion neuropathogenesis and markedly extend survival, even when initiated near clinical signs. These findings should empower development of PrP-lowering therapy for prion disease.


Assuntos
Oligonucleotídeos Antissenso/farmacologia , Oligonucleotídeos Antissenso/uso terapêutico , Doenças Priônicas/tratamento farmacológico , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Descoberta de Drogas , Feminino , Terapia Genética , Camundongos , Camundongos Endogâmicos C57BL , Doenças Priônicas/patologia , Taxa de Sobrevida
5.
Gastroenterology ; 155(6): 1852-1867, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30195448

RESUMO

BACKGROUND & AIMS: Loss of claudin 18 (CLDN18), a membrane-spanning tight junction protein, occurs during early stages of development of gastric cancer and associates with shorter survival times of patients. We investigated whether loss of CLDN18 occurs in mice that develop intraepithelial neoplasia with invasive glands due to infection with Helicobacter pylori, and whether loss is sufficient to promote the development of similar lesions in mice with or without H pylori infection. METHODS: We performed immunohistochemical analyses in levels of CLDN18 in archived tissues from B6:129 mice infected with H pylori for 6 to 15 months. We analyzed gastric tissues from B6:129S5-Cldn18tm1Lex/Mmucd mice, in which the CLDN18 gene was disrupted in gastric tissues (CLDN18-knockout mice), or from control mice with a full-length CLDN18 gene (CLDN18+/+; B6:129S5/SvEvBrd) or heterozygous disruption of CLDN18 (CLDN18+/-; B6:129S5/SvEvBrd) that were infected with H pylori SS1 or PMSS1 at 6 weeks of age and tissues collected for analysis at 20 and 30 weeks after infection. Tissues from CLDN18-knockout mice and control mice with full-length CLDN18 gene expression were also analyzed without infection at 7 weeks and 2 years after birth. Tissues from control and CLDN18-knockout mice were analyzed by electron microscopy, stained by conventional methods and analyzed for histopathology, prepared by laser capture microdissection and analyzed by RNAseq, and immunostained for lineage markers, proliferation markers, and stem cell markers and analyzed by super-resolution or conventional confocal microscopy. RESULTS: CLDN18 had a basolateral rather than apical tight junction localization in gastric epithelial cells. B6:129 mice infected with H pylori, which developed intraepithelial neoplasia with invasive glands, had increasing levels of CLDN18 loss over time compared with uninfected mice. In B6:129 mice infected with H pylori compared with uninfected mice, CLDN18 was first lost from most gastric glands followed by disrupted and reduced expression in the gastric neck and in surface cells. Gastric tissues from CLDN18-knockout mice had low levels of inflammation but increased cell proliferation, expressed markers of intestinalized proliferative spasmolytic polypeptide-expressing metaplasia, and had defects in signal transduction pathways including p53 and STAT signaling by 7 weeks after birth compared with full-length CLDN18 gene control mice. By 20 to 30 weeks after birth, gastric tissues from uninfected CLDN18-knockout mice developed intraepithelial neoplasia that invaded the submucosa; by 2 years, gastric tissues contained large and focally dysplastic polypoid tumors with invasive glands that invaded the serosa. CONCLUSIONS: H pylori infection of B6:129 mice reduced the expression of CLDN18 early in gastric cancer progression, similar to previous observations from human gastric tissues. CLDN18 regulates cell lineage differentiation and cellular signaling in mouse stomach; CLDN18-knockout mice develop intraepithelial neoplasia and then large and focally dysplastic polypoid tumors in the absence of H pylori infection.


Assuntos
Carcinoma in Situ/metabolismo , Claudinas/metabolismo , Infecções por Helicobacter/metabolismo , Neoplasias Gástricas/metabolismo , Animais , Carcinoma in Situ/etiologia , Carcinoma in Situ/microbiologia , Carcinoma in Situ/patologia , Diferenciação Celular , Linhagem da Célula , Progressão da Doença , Feminino , Infecções por Helicobacter/complicações , Helicobacter pylori , Hiperplasia/genética , Hiperplasia/microbiologia , Masculino , Camundongos , Camundongos Knockout , Transdução de Sinais , Estômago/microbiologia , Estômago/patologia , Neoplasias Gástricas/etiologia , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/patologia
6.
PLoS One ; 13(3): e0194443, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29554148

RESUMO

Immune-compromised mouse models allow for testing the preclinical efficacy of human cell transplantations and gene therapy strategies before moving forward to clinical trials. However, CRISPR/Cas9 gene editing of the Wsh/Wsh mouse strain to create an immune-compromised model lacking function of Rag2 and Il2rγ led to unexpected morbidity and mortality. This warranted an investigation to ascertain the cause and predisposing factors associated with the outbreak. Postmortem examination was performed on 15 moribund mice. The main lesions observed in these mice consisted of ascending urogenital tract infections, suppurative otitis media, pneumonia, myocarditis, and meningoencephalomyelitis. As Escherichia coli strains harboring polyketide synthase (pks) genomic island were recently isolated from laboratory mice, the tissue sections from the urogenital tract, heart, and middle ear were subjected to E. coli specific PNA-FISH assay that revealed discrete colonies of E. coli associated with the lesions. Microbiological examination and 16S rRNA sequencing confirmed E. coli-induced infection and septicemia in the affected mice. Further characterization by clb gene analysis and colibactin toxicity assays of the pks+ E. coli revealed colibactin-associated cytotoxicity. Rederivation of the transgenic mice using embryo transfer produced mice with an intestinal flora devoid of pks+ E. coli. Importantly, these barrier-maintained rederived mice have produced multiple litters without adverse health effects. This report is the first to describe acute morbidity and mortality associated with pks+ E. coli urosepsis and meningitis in immunocompromised mice, and highlights the importance of monitoring and exclusion of colibactin-producing pks+ E. coli.


Assuntos
Escherichia coli , Hospedeiro Imunocomprometido , Meningites Bacterianas , Peptídeos/genética , Sepse , Infecções Urinárias , Animais , Escherichia coli/genética , Escherichia coli/imunologia , Escherichia coli/isolamento & purificação , Meningites Bacterianas/genética , Meningites Bacterianas/imunologia , Meningites Bacterianas/microbiologia , Camundongos , Camundongos Transgênicos , Peptídeos/imunologia , Policetídeos/imunologia , Sepse/genética , Sepse/imunologia , Sepse/microbiologia , Infecções Urinárias/genética , Infecções Urinárias/imunologia , Infecções Urinárias/microbiologia
7.
Comp Med ; 68(1): 25-30, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29460718

RESUMO

A laboratory-housed, wild-caught, subadult, male meadow jumping mouse (Zapus hudsonius) presented with extensive scaling of the face, limbs, and tail and severe edema of the paws. Postmortem examination revealed marked distal limb edema with focal digital hematomas and white scales, scabs, and crusts affecting the majority of nonhaired skin. Histopathologic analysis revealed severe, multifocal, chronic-active exudative and proliferative dermatitis characterized by multilaminated crusts covering the epidermis. The epidermis was expanded by hyperkeratosis, acanthosis, and hyperplasia. The superficial dermis contained moderate edema, hemorrhage, and pigmentary incontinence, and was infiltrated by granulocytes and mononuclear cells. The laminated crusts contained numerous branching filaments of gram-positive coccoid bodies arranged in parallel rows, consistent with cutaneous Dermatophilus congolensis infection. This diagnosis was confirmed through bacterial culture and 16S rRNA PCR analysis. In the presented case, factors that might have contributed to disease progression include climatic conditions at the capture site and stress associated with trapping and laboratory housing.


Assuntos
Dermatite/veterinária , Doenças dos Roedores/microbiologia , Roedores , Dermatopatias Bacterianas/veterinária , Actinobacteria/isolamento & purificação , Animais , Dermatite/microbiologia , Dermatite/patologia , Masculino , Dermatopatias Bacterianas/microbiologia
8.
J Am Assoc Lab Anim Sci ; 56(6): 802-806, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29256376

RESUMO

Although zebra finches (Taeniopygia guttata) have been used in biomedical research for many years, no published reports are available about euthanizing these small birds. In this study, we compared 5 methods for zebra finch euthanasia: sodium pentobarbital (NaP) given intracoelomically with physical restraint but no anesthesia; isoflurane anesthesia followed by intracoelomic injection of NaP; and CO2 asphyxiation at 20%, 40%, and 80% chamber displacement rates (percentage of chamber volume per minute). Birds undergoing euthanasia were videorecorded and scored by 2 observers for behaviors potentially related to discomfort or distress. Time to recumbency and time until respiratory arrest (RA) were also assessed. RA was achieved faster by using NaP in a conscious bird compared to using isoflurane anesthesia followed by NaP; however, neither method caused behaviors that might affect animal welfare, such as open-mouth breathing, to any appreciable extent. Among the CO2 treatment groups, there was an inverse correlation between the chamber displacement rate used and the duration of open-mouth breathing, onset of head retroflexion, and time to RA. The results demonstrate that the intracoelomic administration of NaP in an awake, restrained zebra finch is a rapid and effective method of euthanasia. If CO2 is used to euthanize these birds, a high displacement rate (for example, 80%) will minimize the duration of the procedure and associated behaviors.


Assuntos
Bem-Estar do Animal , Dióxido de Carbono/administração & dosagem , Eutanásia Animal/métodos , Tentilhões/fisiologia , Isoflurano/administração & dosagem , Pentobarbital/administração & dosagem , Animais , Animais de Laboratório , Feminino , Tentilhões/classificação , Masculino
9.
World J Gastroenterol ; 21(40): 11411-27, 2015 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-26523106

RESUMO

Long-term chronic infection with Helicobacter pylori (H. pylori) is a risk factor for gastric cancer development. In the multi-step process that leads to gastric cancer, tight junction dysfunction is thought to occur and serve as a risk factor by permitting the permeation of luminal contents across an otherwise tight mucosa. Mechanisms that regulate tight junction function and structure in the normal stomach, or dysfunction in the infected stomach, however, are largely unknown. Although conventional tight junction components are expressed in gastric epithelial cells, claudins regulate paracellular permeability and are likely the target of inflammation or H. pylori itself. There are 27 different claudin molecules, each with unique properties that render the mucosa an intact barrier that is permselective in a way that is consistent with cell physiology. Understanding the architecture of tight junctions in the normal stomach and then changes that occur during infection is important but challenging, because most of the reports that catalog claudin expression in gastric cancer pathogenesis are contradictory. Furthermore, the role of H. pylori virulence factors, such as cytotoxin-associated gene A and vacoulating cytotoxin, in regulating tight junction dysfunction during infection is inconsistent in different gastric cell lines and in vivo, likely because non-gastric epithelial cell cultures were initially used to unravel the details of their effects on the stomach. Hampering further study, as well, is the relative lack of cultured cell models that have tight junction claudins that are consistent with native tissues. This summary will review the current state of knowledge about gastric tight junctions, normally and in H. pylori infection, and make predictions about the consequences of claudin reorganization during H. pylori infection.


Assuntos
Absorção Gástrica , Mucosa Gástrica/microbiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/patogenicidade , Junções Íntimas/microbiologia , Animais , Mucosa Gástrica/metabolismo , Helicobacter pylori/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Permeabilidade , Proteínas de Junções Íntimas/metabolismo , Junções Íntimas/metabolismo , Virulência , Fatores de Virulência/metabolismo
10.
Blood ; 117(19): 5112-22, 2011 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-21403126

RESUMO

Under persistent antigenic stimulation, virus-specific CD8⁺ T cells become increasingly dysfunctional and up-regulate several inhibitory molecules such as killer lectin-like receptor G1 (KLRG1). Here, we demonstrate that HIV-1 antigen-specific T cells from subjects with chronic-progressive HIV-1 infection have significantly elevated KLRG1 expression (P < .001); show abnormal distribution of E-cadherin, the natural ligand of KLRG1, in the intestinal mucosa; and have elevated levels of systemic soluble E-cadherin (sE-cadherin) that significantly correlate with HIV-1 viral load (R = 0.7, P = .004). We furthermore demonstrate that in the presence of sE-cadherin, KLRG1(hi) HIV-1-specific CD8⁺ T cells are impaired in their ability to respond by cytokine secretion on antigenic stimulation (P = .002) and to inhibit viral replication (P = .03) in vitro. Thus, these data suggest a critical mechanism by which the disruption of the intestinal epithelium associated with HIV-1 leads to increased systemic levels of sE-cadherin, which inhibits the effector functions of KLRG1(hi)-expressing HIV-1-specific CD8⁺ T cells systemically.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Caderinas/metabolismo , Infecções por HIV/imunologia , Lectinas Tipo C/biossíntese , Transativadores/biossíntese , Linfócitos T CD8-Positivos/metabolismo , Caderinas/imunologia , Moléculas de Adesão Celular/imunologia , Moléculas de Adesão Celular/metabolismo , Separação Celular , Colo/imunologia , Colo/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Imunofluorescência , Infecções por HIV/metabolismo , HIV-1/imunologia , Humanos , Imuno-Histoquímica , Lectinas Tipo C/imunologia , Ativação Linfocitária/imunologia , Masculino , Receptores Imunológicos , Transativadores/imunologia , Replicação Viral/imunologia
11.
Brain ; 134(Pt 2): 534-41, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21216828

RESUMO

In the central nervous system of patients with multiple sclerosis, B cell aggregates populate the meninges, raising the central question as to whether these structures relate to the B cell infiltrates found in parenchymal lesions or instead, represent a separate central nervous system immune compartment. We characterized the repertoires derived from meningeal B cell aggregates and the corresponding parenchymal infiltrates from brain tissue derived primarily from patients with progressive multiple sclerosis. The majority of expanded antigen-experienced B cell clones derived from meningeal aggregates were also present in the parenchyma. We extended this investigation to include 20 grey matter specimens containing meninges, 26 inflammatory plaques, 19 areas of normal appearing white matter and cerebral spinal fluid. Analysis of 1833 B cell receptor heavy chain variable region sequences demonstrated that antigen-experienced clones were consistently shared among these distinct compartments. This study establishes a relationship between extraparenchymal lymphoid tissue and parenchymal infiltrates and defines the arrangement of B cell clones that populate the central nervous system of patients with multiple sclerosis.


Assuntos
Linfócitos B/imunologia , Sistema Nervoso Central/imunologia , Células Clonais/imunologia , Meninges/imunologia , Esclerose Múltipla/imunologia , Humanos , Região Variável de Imunoglobulina/imunologia
12.
Development ; 137(13): 2147-56, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20530543

RESUMO

Coactivator-associated arginine methyltransferase I (CARM1; PRMT4) regulates gene expression by multiple mechanisms including methylation of histones and coactivation of steroid receptor transcription. Mice lacking CARM1 are small, fail to breathe and die shortly after birth, demonstrating the crucial role of CARM1 in development. In adults, CARM1 is overexpressed in human grade-III breast tumors and prostate adenocarcinomas, and knockdown of CARM1 inhibits proliferation of breast and prostate cancer cell lines. Based on these observations, we hypothesized that loss of CARM1 in mouse embryos would inhibit pulmonary cell proliferation, resulting in respiratory distress. By contrast, we report here that loss of CARM1 results in hyperproliferation of pulmonary epithelial cells during embryonic development. The lungs of newborn mice lacking CARM1 have substantially reduced airspace compared with their wild-type littermates. In the absence of CARM1, alveolar type II cells show increased proliferation. Electron microscopic analyses demonstrate that lungs from mice lacking CARM1 have immature alveolar type II cells and an absence of alveolar type I cells. Gene expression analysis reveals a dysregulation of cell cycle genes and markers of differentiation in the Carm1 knockout lung. Furthermore, there is an overlap in gene expression in the Carm1 knockout and the glucocorticoid receptor knockout lung, suggesting that hyperproliferation and lack of maturation of the alveolar cells are at least in part caused by attenuation of glucocorticoid-mediated signaling. These results demonstrate for the first time that CARM1 inhibits pulmonary cell proliferation and is required for proper differentiation of alveolar cells.


Assuntos
Células Epiteliais/metabolismo , Pulmão/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Animais , Proliferação de Células , Células Endoteliais/metabolismo , Glucocorticoides/metabolismo , Camundongos , Alvéolos Pulmonares/metabolismo , Transcrição Gênica
13.
Am J Surg Pathol ; 34(6): 882-91, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20442643

RESUMO

Nearly 100% of Burkitt lymphomas (BLs) and 5% to 8% of diffuse large B-cell lymphomas (DLBCLs) harbor a balanced translocation involving c-MYC. Although characteristic morphologic and immunophenotypic features can identify BL in most cases, tumors with atypical features are often encountered in clinical practice. Furthermore, no morphologic or immunophenotypic finding can predict an underlying c-MYC translocation in DLBCL with certainty. Here we report on a novel monoclonal antibody recognizing the c-myc protein in formalin-fixed, paraffin-embedded tissue which we used to evaluate a spectrum of aggressive B-cell lymphomas by standard immunohistochemistry. Cases consisted of 17 BLs (15 cases with confirmed c-MYC translocation), 19 DLBCLs without a c-MYC translocation, 5 DLBCLs with a c-MYC translocation, and 2 B-cell lymphomas, unclassifiable, with features intermediate between DLBCL and BL (intermediate DBLCL/BL, one case with c-MYC translocation and one case without a c-MYC translocation). The intensity and subcellular localization of tumor-specific staining for c-myc protein was determined independently by 2 pathologists and in a blinded fashion for each case. We observed c-myc expression in the tumor cells of all cases regardless of c-MYC status. Among BLs, c-myc protein primarily localized to the nucleus of tumor cells in 15 of 17 cases (88%) and equally localized to the nucleus and cytoplasm of tumor cells in 2 of 17 cases (12%). In no case did c-myc protein primarily localize to the cytoplasm. In contrast, among DLBCLs lacking a c-MYC translocation the c-myc protein primarily localized to the cytoplasm of the tumor cells in 18 of 19 cases (95%) and equally localized to the nucleus and cytoplasm in the tumor cells in 1 of 19 cases (5%). In no case did c-myc protein primarily localize to the nucleus. Among DLBCLs with a c-MYC translocation and intermediate DBLCL/BLs, the c-myc protein primarily localized to the nucleus, or equally localized to the nucleus and cytoplasm of the tumor cells in 4 of 5 cases (80%) and 2 of 2 cases (100%), respectively. Taken together, we find that a primarily nuclear or mixed nuclear and cytoplasmic staining pattern for c-myc in an aggressive B-cell lymphoma is highly predictive of a c-MYC translocation (positive-predictive value=0.92, negative-predictive value=0.95, P<0.0001). We further show that the subcellular localization of c-myc can be determined with good interobserver agreement among pathologists (kappa statistic=0.90). Thus this novel immunohistochemsitry test is a useful tool for identifying aggressive B-cell lymphomas likely to harbor a c-MYC rearrangement and thus warrant genetic testing.


Assuntos
Anticorpos Monoclonais , Linfoma de Burkitt/genética , Linfoma Difuso de Grandes Células B/genética , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Linfoma de Burkitt/diagnóstico , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Genes myc , Humanos , Imuno-Histoquímica/métodos , Linfoma Difuso de Grandes Células B/diagnóstico , Variações Dependentes do Observador , Translocação Genética
14.
Brain ; 132(Pt 12): 3318-28, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19638446

RESUMO

Multiple sclerosis is an inflammatory demyelinating disease of the central nervous system (CNS) that is thought to be caused by a combination of genetic and environmental factors. To date, considerable evidence has associated Epstein-Barr virus (EBV) infection with disease development. However, it remains controversial whether EBV infects multiple sclerosis brain and contributes directly to CNS immunopathology. To assess whether EBV infection is a characteristic feature of multiple sclerosis brain, a large cohort of multiple sclerosis specimens containing white matter lesions (nine adult and three paediatric cases) with a heterogeneous B cell infiltrate and a second cohort of multiple sclerosis specimens (12 cases) that included B cell infiltration within the meninges and parenchymal B cell aggregates, were examined for EBV infection using multiple methodologies including in situ hybridization, immunohistochemistry and two independent real-time polymerase chain reaction (PCR) methodologies that detect genomic EBV or the abundant EBV encoded RNA (EBER) 1, respectively. We report that EBV could not be detected in any of the multiple sclerosis specimens containing white matter lesions by any of the methods employed, yet EBV was readily detectable in multiple Epstein-Barr virus-positive control tissues including several CNS lymphomas. Furthermore, EBV was not detected in our second cohort of multiple sclerosis specimens by in situ hybridization. However, our real-time PCR methodologies, which were capable of detecting very few EBV infected cells, detected EBV at low levels in only 2 of the 12 multiple sclerosis meningeal specimens examined. Our finding that CNS EBV infection was rare in multiple sclerosis brain indicates that EBV infection is unlikely to contribute directly to multiple sclerosis brain pathology in the vast majority of cases.


Assuntos
Encéfalo/virologia , Infecções por Vírus Epstein-Barr/complicações , Infecções por Vírus Epstein-Barr/patologia , Esclerose Múltipla/patologia , Esclerose Múltipla/virologia , Adulto , Linfócitos B/imunologia , Linfócitos B/virologia , Encéfalo/patologia , Causalidade , Criança , Estudos de Coortes , Infecções por Vírus Epstein-Barr/fisiopatologia , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/imunologia , Humanos , Imunidade Humoral/fisiologia , Células Jurkat , Ativação Linfocitária/imunologia , Esclerose Múltipla/fisiopatologia , Reação em Cadeia da Polimerase/métodos , RNA Viral/análise , RNA Viral/genética , RNA Viral/metabolismo
15.
J Immunol ; 182(5): 3310-7, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19234230

RESUMO

Germ cell tumors are a heterogeneous group of neoplasms derived from residual primordial tissue. These tumors are commonly found in the brain, testes, or ovaries, where they are termed germinomas, seminomas, or dysgerminomas, respectively. Like several other tumor types, germ cell tumors often harbor an immune cell infiltrate that can include substantial numbers of B cells. Yet little is known about whether the humoral immune response affects germ cell tumor biology. To gain a deeper understanding of the role B cells play in this tumor family, we characterized the immune cell infiltrate of all three germ cell tumor subtypes and defined the molecular characteristics of the B cell Ag receptor expressed by tumor-associated B cells. Immunohistochemistry revealed a prominent B cell infiltrate in the microenvironment of all tumors examined and clear evidence of extranodal lymphoid follicles with germinal center-like architecture in a subset of specimens. Molecular characterization of the Ig variable region from 320 sequences expressed by germ cell tumor-infiltrating B cells revealed clear evidence of Ag experience, in that the cardinal features of an Ag-driven B cell response were present: significant somatic mutation, isotype switching, and codon insertion/deletion. This characterization also revealed the presence of both B cell clonal expansion and variation, suggesting that local B cell maturation most likely occurs within the tumor microenvironment. In contrast, sequences from control tissues and peripheral blood displayed none of these characteristics. Collectively, these data strongly suggest that an adaptive and specific humoral immune response is occurring within the tumor microenvironment.


Assuntos
Anticorpos Antineoplásicos/biossíntese , Antígenos de Neoplasias/imunologia , Neoplasias Embrionárias de Células Germinativas/imunologia , Neoplasias Embrionárias de Células Germinativas/metabolismo , Anticorpos Antineoplásicos/genética , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Subpopulações de Linfócitos B/patologia , Movimento Celular/imunologia , Células Clonais , Disgerminoma/imunologia , Disgerminoma/metabolismo , Disgerminoma/patologia , Germinoma/imunologia , Germinoma/metabolismo , Germinoma/patologia , Humanos , Cadeias Pesadas de Imunoglobulinas/biossíntese , Cadeias Pesadas de Imunoglobulinas/genética , Região Variável de Imunoglobulina/biossíntese , Região Variável de Imunoglobulina/genética , Meduloblastoma/imunologia , Meduloblastoma/metabolismo , Meduloblastoma/patologia , Dados de Sequência Molecular , Neoplasias Embrionárias de Células Germinativas/patologia , Seminoma/imunologia , Seminoma/metabolismo , Seminoma/patologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...