Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Nature ; 457(7230): 722-5, 2009 Feb 05.
Article in English | MEDLINE | ID: mdl-18978771

ABSTRACT

Natural killer (NK) cells are classically viewed as lymphocytes that provide innate surveillance against virally infected cells and tumour cells through the release of cytolytic mediators and interferon (IFN)-gamma. In humans, blood CD56(dim) NK cells specialize in the lysis of cell targets. In the lymph nodes, CD56(bright) NK cells secrete IFN-gamma cooperating with dendritic cells and T cells in the generation of adaptive responses. Here we report the characterization of a human NK cell subset located in mucosa-associated lymphoid tissues, such as tonsils and Peyer's patches, which is hard-wired to secrete interleukin (IL)-22, IL-26 and leukaemia inhibitory factor. These NK cells, which we refer to as NK-22 cells, are triggered by acute exposure to IL-23. In vitro, NK-22-secreted cytokines stimulate epithelial cells to secrete IL-10, proliferate and express a variety of mitogenic and anti-apoptotic molecules. NK-22 cells are also found in mouse mucosa-associated lymphoid tissues and appear in the small intestine lamina propria during bacterial infection, suggesting that NK-22 cells provide an innate source of IL-22 that may help constrain inflammation and protect mucosal sites.


Subject(s)
Immunity, Mucosal/immunology , Interleukins/biosynthesis , Interleukins/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Animals , CD56 Antigen/analysis , Cell Adhesion , Chemokine CCL20/biosynthesis , Chemokine CCL20/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Interleukin-10/metabolism , Interleukin-23/immunology , Interleukins/metabolism , Killer Cells, Natural/metabolism , Leukemia Inhibitory Factor/biosynthesis , Mice , Monocytes/immunology , Palatine Tonsil/cytology , Palatine Tonsil/immunology , Receptors, CCR6/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Toll-Like Receptors/immunology , Interleukin-22
2.
Physiol Genomics ; 45(15): 667-83, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23715263

ABSTRACT

In a screen for genes expressed specifically in gastric mucous neck cells, we identified GKN3, the recently discovered third member of the gastrokine family. We present confirmatory mouse data and novel porcine data showing that mouse GKN3 expression is confined to mucous cells of the corpus neck and antrum base and is prominently expressed in metaplastic lesions. GKN3 was proposed originally to be expressed in some human populations and a pseudogene in others. To investigate that hypothesis, we studied human GKN3 evolution in the context of its paralogous genomic neighbors, GKN1 and GKN2. Haplotype analysis revealed that GKN3 mimics GKN2 in patterns of exonic SNP allocation, whereas GKN1 appeared to be more stringently selected. GKN3 showed signatures of both directional selection and population based selective sweeps in humans. One such selective sweep includes SNP rs10187256, originally identified as an ancestral tryptophan to premature STOP codon mutation. The derived (nonancestral) allele went to fixation in Asia. We show that another SNP, rs75578132, identified 5 bp downstream of rs10187256, exhibits a second selective sweep in almost all Europeans, some Latinos, and some Africans, possibly resulting from a reintroduction of European genes during African colonization. Finally, we identify a mutation that would destroy the splice donor site in the putative exon3-intron3 boundary, which occurs in all human genomes examined to date. Our results highlight a stomach-specific human genetic locus, which has undergone various selective sweeps across European, Asian, and African populations and thus reflects geographic and ethnic patterns in genome evolution.


Subject(s)
Carrier Proteins/genetics , Evolution, Molecular , Genetic Loci/genetics , Membrane Proteins/genetics , Pseudogenes/genetics , Racial Groups/genetics , Selection, Genetic/genetics , Animals , Carrier Proteins/metabolism , Computational Biology , DNA Primers/genetics , Fluorescent Antibody Technique , Gastric Mucosa/metabolism , Genetics, Population , Genotype , Haplotypes/genetics , Humans , Likelihood Functions , Macaca mulatta/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL/genetics , Microarray Analysis , Microscopy, Confocal , Models, Genetic , Mutation/genetics , Phylogeny , Polymorphism, Single Nucleotide/genetics , Species Specificity , Sus scrofa/genetics
3.
Am J Pathol ; 179(2): 1015-29, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21704007

ABSTRACT

Cirrhotic septa harbor vessels and inflammatory, fibrogenic, and ductular epithelial cells, collectively referred to as the ductular reaction (DR). Lack of the DR in the stromal compartment around hepatocellular carcinoma (HCC) has been documented; however, the relationship of epithelial keratin 19 (K19) structures to progression of intralesional carcinogenesis has not been explored. K19 immunoreactivity in the stromal compartment around 176 nodules in cirrhotic explants was examined. Quantitative differences (P < 0.0001) were manifested in three distinct histologically identifiable patterns: "complex" around cirrhotic nodules (CN), "attenuated" around dysplastic nodules (DN), and "absent" around HCC. Markers of necrosis or apoptosis could not explain the perinodular K19 epithelial loss; however, multicolor immunolabeling for K19, vimentin, E-Cadherin, SNAIL, and fibroblast-specific protein 1 (FSP-1) demonstrated discrepancies in immunophenotype and cytomorphologic features. Variability of cellular features was accompanied by an overall decrease in epithelial markers and significantly increased fractions of SNAIL- and FSP-1-positive cells in the DR around DN when compared with CN (P < 0.0001). Immunolabeling of transforming growth factor-ß signaling components (TGFßR1, SMAD3, and pSMAD2/3) demonstrated increased percentages of pSMAD2/3 around DN when compared with CN (P < 0.0001). These findings collectively suggest marked alterations in cellular identity as an underlying mechanism for the reproducible extralesional K19 pattern that parallels progressive stages of intranodular hepatocarcinogenesis. Paracrine signaling is proposed as a link that emphasizes the importance of the epithelial-stromal compartment in malignant progression of HCC in cirrhosis.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Epithelium/metabolism , Gene Expression Regulation, Neoplastic , Gene Expression Regulation , Keratin-19/metabolism , Liver Cirrhosis/metabolism , Liver Neoplasms/metabolism , Apoptosis , Biopsy , Humans , Models, Biological , Necrosis , Predictive Value of Tests , Signal Transduction , Transforming Growth Factor beta/metabolism
4.
Physiol Genomics ; 43(3): 174-86, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21098683

ABSTRACT

Despite their divergent developmental ancestry, plasma cells and gastric zymogenic (chief) cells share a common function: high-capacity secretion of protein. Here we show that both cell lineages share increased expression of a cassette of 269 genes, most of which regulate endoplasmic reticulum (ER) and Golgi function, and they both induce expression of the transcription factors X-box binding protein 1 (Xbp1) and Mist1 during terminal differentiation. XBP1 is known to augment plasma cell function by establishing rough ER, and MIST1 regulates secretory vesicle trafficking in zymogenic cells. We examined morphology and function of plasma cells in wild-type and Mist1(-/-) mice and found subtle differences in ER structure but no overall defect in plasma cell function, suggesting that Mist1 may function redundantly in plasma cells. We next reasoned that MIST1 might be useful as a novel and reliable marker of plasma cells. We found that MIST1 specifically labeled normal plasma cells in mouse and human tissues, and, moreover, its expression was also characteristic of plasma cell differentiation in a cohort of 12 human plasma cell neoplasms. Overall, our results show that MIST1 is enriched upon plasma cell differentiation as a part of a genetic program facilitating secretory cell function and also that MIST1 is a novel marker of normal and neoplastic plasma cells in mouse and human tissues.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Plasma Cells/cytology , Plasma Cells/metabolism , Transcription Factors/metabolism , Animals , Antibodies/metabolism , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Chief Cells, Gastric/cytology , Chief Cells, Gastric/drug effects , Chief Cells, Gastric/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Flow Cytometry , Gene Expression Regulation/drug effects , Humans , Intracellular Membranes/metabolism , Leukemia, Plasma Cell/metabolism , Leukemia, Plasma Cell/pathology , Lipopolysaccharides/pharmacology , Mice , Plasma Cells/drug effects , Promoter Regions, Genetic/genetics , Regulatory Factor X Transcription Factors , Syndecan-1/metabolism , Transcription Factors/genetics , X-Box Binding Protein 1
5.
Am J Pathol ; 177(3): 1514-33, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20709804

ABSTRACT

The lack of reliable molecular markers for normal differentiated epithelial cells limits understanding of human gastric carcinogenesis. Recognized precursor lesions for gastric adenocarcinoma are intestinal metaplasia and spasmolytic polypeptide expressing metaplasia (SPEM), defined here by ectopic CDX2 and TFF2 expression, respectively. In mice, expression of the bHLH transcription factor MIST1, normally restricted to mature chief cells, is down-regulated as chief cells undergo experimentally induced metaplasia. Here, we show MIST1 expression is also a specific marker of human chief cells. SPEM, with and without MIST1, is present in human lesions and, akin to murine data, likely represents transitional (TFF2(+)/MIST1(+) = "hybrid"-SPEM) and established (TFF2(+)/MIST1(-) = SPEM) stages. Co-visualization of MIST1 and CDX2 shows similar progressive loss of MIST1 with a transitional, CDX2(+)/MIST1(-) hybrid-intestinal metaplasia stage. Interinstitutional analysis and comparison of findings in tissue microarrays, resection specimens, and biopsies (n > 400 samples), comprising the entire spectrum of recognized stages of gastric carcinogenesis, confirm MIST1 expression is restricted to the chief cell compartment in normal oxyntic mucosa, rare in established metaplastic lesions, and lost in intraepithelial neoplasia/dysplasia and carcinoma of various types with the exception of rare chief cell carcinoma ( approximately 1%). Our findings implicate MIST1 as a reliable marker of mature, healthy chief cells, and we provide the first evidence that metaplasia in humans arises at least in part from the chief cell lineage.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Chief Cells, Gastric/metabolism , Gastrointestinal Diseases/metabolism , Gastrointestinal Tract/metabolism , Adaptor Proteins, Signal Transducing/genetics , Blotting, Western , Cells, Cultured , Chief Cells, Gastric/pathology , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gastrointestinal Diseases/genetics , Gastrointestinal Diseases/pathology , Gastrointestinal Tract/pathology , Humans , Immunohistochemistry , Metaplasia/genetics , Metaplasia/metabolism , Metaplasia/pathology , Microscopy, Confocal , Tissue Array Analysis , Trefoil Factor-2
6.
Open Forum Infect Dis ; 8(2): ofaa631, 2021 Feb.
Article in English | MEDLINE | ID: mdl-34853795

ABSTRACT

BACKGROUND: Amid the enduring pandemic, there is an urgent need for expanded access to rapid, sensitive, and inexpensive coronavirus disease 2019 (COVID-19) testing worldwide without specialized equipment. We developed a simple test that uses colorimetric reverse transcription loop-mediated isothermal amplification (RT-LAMP) to detect severe acute resrpiratory syndrome coronavirus 2 (SARS-CoV-2) in 40 minutes from sample collection to result. METHODS: We tested 135 nasopharyngeal specimens from patients evaluated for COVID-19 infection at Massachusetts General Hospital. Specimens were either added directly to RT-LAMP reactions, inactivated by a combined chemical and heat treatment step, or inactivated then purified with a silica particle-based concentration method. Amplification was performed with 2 SARS-CoV-2-specific primer sets and an internal specimen control; the resulting color change was visually interpreted. RESULTS: Direct RT-LAMP testing of unprocessed specimens could only reliably detect samples with abundant SARS-CoV-2 (>3 000 000 copies/mL), with sensitivities of 50% (95% CI, 28%-72%) and 59% (95% CI, 43%-73%) in samples collected in universal transport medium and saline, respectively, compared with quantitative polymerase chain reaction (qPCR). Adding an upfront RNase inactivation step markedly improved the limit of detection to at least 25 000 copies/mL, with 87.5% (95% CI, 72%-95%) sensitivity and 100% specificity (95% CI, 87%-100%). Using both inactivation and purification increased the assay sensitivity by 10-fold, achieving a limit of detection comparable to commercial real-time PCR-based diagnostics. CONCLUSIONS: By incorporating a fast and inexpensive sample preparation step, RT-LAMP accurately detects SARS-CoV-2 with limited equipment for about US$6 per sample, making this a potentially ideal assay to increase testing capacity, especially in resource-limited settings.

8.
Am J Surg Pathol ; 36(11): 1709-17, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23073328

ABSTRACT

The management of hereditary diffuse-type gastric cancer revolves around surveillance biopsies and the timing of prophylactic gastrectomy. In the absence of a validated surveillance biopsy protocol, we modeled bioptic diagnostic yield on the basis of the topographic distribution of cancer foci in a series of 10 gastrectomies in CDH1-mutation carriers. Complete histologic examination was performed in all cases, and 1817 slides were evaluated for the presence of in situ, intramucosal, or submucosal diffuse-type carcinoma. Detailed maps determined the density of cancer foci. On the basis of the number of sampled glands per biopsy in routine surveillance preoperative endoscopy, we estimated the theoretical number of biopsies necessary for a 90% rate of detection of neoplastic foci, and we evaluated this number, taking into account the regional distribution of these foci. A total of 96 m of gastric mucosa with ≈ 1,193,453 gastric glands yielded 302 cancer foci [in situ (n=89), intramucosal (n=209), and submucosal (n=4)] spanning the width of a total of 1820 glands (8 to 1205 per case; average 182 ± 115). On the basis of the number of glands per stomach and the average number of glands sampled during surveillance biopsy (28.7 ± 1.7; range, 0 to 79; n=112), the theoretical number of biopsies necessary to capture at least 1 cancer focus was estimated to be 1768 (range, 50 to 5832) to assure a 90% detection rate. Mapping of cancer foci showed the highest density in the anterior proximal fundus (37%) and cardia/proximal fundus (27%). Our results argue for the incorporation of cancer focus distribution into any biopsy protocol, although detection is likely to remain extremely low, and they call into question the validity of endoscopic surveillance.


Subject(s)
Adenocarcinoma/diagnosis , Cadherins/genetics , Carcinoma in Situ/diagnosis , Gastroscopy/methods , Mutation , Stomach Neoplasms/diagnosis , Adenocarcinoma/genetics , Adenocarcinoma/surgery , Adult , Antigens, CD , Biopsy , Carcinoma in Situ/genetics , Carcinoma in Situ/surgery , Cardia/pathology , Female , Gastrectomy , Gastric Fundus/pathology , Gastric Mucosa/pathology , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Neoplastic Syndromes, Hereditary/diagnosis , Neoplastic Syndromes, Hereditary/genetics , Population Surveillance/methods , Retrospective Studies , Stomach Neoplasms/genetics , Stomach Neoplasms/surgery
9.
Am J Surg Pathol ; 33(6): 835-43, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19092631

ABSTRACT

Mucoepidermoid carcinoma (MEC) of the uterine cervix is a controversial entity. By strict morphologic criteria, the tumor has features identical to those of salivary gland MEC and is characterized by nests composed of 3 cell types (epidermoid, intermediate, and mucin producing) in the absence of overt glandular differentiation. Nonetheless, the entity is not recognized in the current World Health Organization classification of cervical tumors. Given the morphologic similarity between MEC of the cervix and MEC of the salivary glands, we sought to determine if MEC of the cervix harbors the t(11;19)(q21;p13) characteristic of MEC of the major and minor salivary glands, a rearrangement that results in fusion of the cyclic adenosine 3',5' monophosphate coactivator CRTC1 to the Notch coactivator MAML2. We identified 7 cervical tumors from our departmental files and performed reverse transcription-polymerase chain reaction and fluorescence in situ hybridization-based molecular analysis for rearrangements of CRTC1 and MAML2; 14 conventional cervical adenosquamous carcinomas were used as controls. Analysis of the cervical MECs demonstrated a CRTC1-MAML2 fusion in 1 case, rearrangements of CRTC1 in 4 cases, and aberrations of MAML2 in 5 cases (rearrangements in 2 cases, amplification in 3 cases). All MEC showed aberrations of at least 1 of the loci, whereas none of the cervical adenosquamous carcinomas harbored rearrangements or amplification of either locus. Our results demonstrate that cervical tumors defined as MEC by strict morphologic criteria harbor genetic aberrations involving the genes characteristically rearranged in MEC of the salivary glands, and suggest that cervical MEC is an entity distinct from conventional cervical adenosquamous carcinoma. The development of drug therapy targeted to the genes rearranged in MEC underscores the importance of correct classification of cervical MEC because the diagnosis may hold therapeutic implications different from other cervical malignancies.


Subject(s)
Carcinoma, Mucoepidermoid/pathology , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Oncogene Proteins, Fusion/genetics , Transcription Factors/genetics , Uterine Cervical Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Carcinoma, Mucoepidermoid/genetics , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 19 , Female , Gene Amplification , Gene Rearrangement , Humans , In Situ Hybridization, Fluorescence , Middle Aged , Neoplasm Staging , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators , Uterine Cervical Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL