RESUMO
A predominantly diffuse growth pattern and CD23 co-expression are uncommon findings in nodal follicular lymphoma and can create diagnostic challenges. A single case series in 2009 (Katzenberger et al) proposed a unique morphologic variant of nodal follicular lymphoma, characterized by a predominantly diffuse architecture, lack of the t(14;18) IGH/BCL2 translocation, presence of 1p36 deletion, frequent inguinal lymph node involvement, CD23 co-expression, and low clinical stage. Other studies on CD23+ follicular lymphoma, while associating inguinal location, have not specifically described this architecture. In addition, no follow-up studies have correlated the histopathologic and cytogenetic/molecular features of these cases, and they remain a diagnostic problem. We identified 11 cases of diffuse, CD23+ follicular lymphoma with histopathologic features similar to those described by Katzenberger et al. Along with pertinent clinical information, we detail their histopathology, IGH/BCL2 translocation status, lymphoma-associated chromosomal gains/losses, and assessment of mutations in 220 lymphoma-associated genes by massively parallel sequencing. All cases showed a diffuse growth pattern around well- to ill-defined residual germinal centers, uniform CD23 expression, mixed centrocytic/centroblastic cytology, and expression of at least one germinal center marker. Ten of 11 involved inguinal lymph nodes, 5 solely. By fluorescence in situ hybridization analysis, the vast majority lacked IGH/BCL2 translocation (9/11). Deletion of 1p36 was observed in five cases and included TNFRSF14. Of the six cases lacking 1p36 deletion, TNFRSF14 mutations were identified in three, highlighting the strong association of 1p36/TNFRSF14 abnormalities with this follicular lymphoma variant. In addition, 9 of the 11 cases tested (82%) had STAT6 mutations and nuclear P-STAT6 expression was detectable in the mutated cases by immunohistochemistry. The proportion of STAT6 mutations is higher than recently reported in conventional follicular lymphoma (11%). These findings lend support for a clinicopathologic variant of t(14;18) negative nodal follicular lymphoma and suggests importance of the interleukin (IL)-4/JAK/STAT6 pathway in this variant.
Assuntos
Biomarcadores Tumorais/genética , Transtornos Cromossômicos/genética , Cromossomos Humanos Par 14 , Cromossomos Humanos Par 18 , Linfoma Folicular/genética , Mutação , Receptores de IgE/análise , Membro 14 de Receptores do Fator de Necrose Tumoral/genética , Fator de Transcrição STAT6/genética , Translocação Genética , Adulto , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/análise , Deleção Cromossômica , Transtornos Cromossômicos/imunologia , Transtornos Cromossômicos/patologia , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 1/imunologia , Análise Mutacional de DNA/métodos , Feminino , Genes de Cadeia Pesada de Imunoglobulina , Predisposição Genética para Doença , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Linfoma Folicular/química , Linfoma Folicular/imunologia , Linfoma Folicular/patologia , Masculino , Pessoa de Meia-Idade , Fenótipo , Fosforilação , Proteínas Proto-Oncogênicas c-bcl-2/genética , Fator de Transcrição STAT6/análiseRESUMO
CS1 (CRACC, CD319) and 2B4 (CD244), members of the signalling lymphocyte activation molecule (SLAM) family receptors, regulate various immune functions. Genes encoding SLAM family receptors are located at 1q23, implicated in systemic lupus erythematosus (SLE). In this study, we have investigated the expression and alternative splicing of CS1 and 2B4 in immune cells from SLE patients. The surface expression of CS1 and 2B4 on total peripheral blood mononuclear cells (PBMCs), T, B, natural killer (NK) cells and monocytes in 45 patients with SLE and 30 healthy individuals was analysed by flow cytometry. CS1-positive B cell population was increased significantly in SLE patients. Because CS1 is a self-ligand and homophilic interaction of CS1 induces B cell proliferation and autocrine cytokine secretion, this could account for autoreactive B cell proliferation in SLE. The proportion of NK cells and monocytes expressing 2B4 on their surface was significantly lower in patients with SLE compared to healthy controls. Our study demonstrated altered expression of splice variants of CS1 and 2B4 that mediate differential signalling in PBMC from patients with SLE.
Assuntos
Processamento Alternativo/imunologia , Antígenos CD/imunologia , Leucócitos Mononucleares/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Receptores Imunológicos/imunologia , Transdução de Sinais/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos CD/biossíntese , Comunicação Autócrina/imunologia , Estudos de Casos e Controles , Proliferação de Células , Cromossomos Humanos Par 1/imunologia , Cromossomos Humanos Par 1/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Feminino , Citometria de Fluxo , Humanos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Lúpus Eritematoso Sistêmico/metabolismo , Lúpus Eritematoso Sistêmico/patologia , Masculino , Pessoa de Meia-Idade , Receptores Imunológicos/biossíntese , Família de Moléculas de Sinalização da Ativação LinfocitáriaRESUMO
The filaggrin (FLG) gene is one of the most widely replicated susceptibility genes for atopic dermatitis (AD) so far. Yet, FLG mutations cannot fully account for the original linkage peak on chromosome 1q21, a region comprising the so-called epidermal differentiation complex (EDC). Since the EDC contains numerous genes relevant for epidermal differentiation, we sought to evaluate variation in other genes located in this region in a German AD case-control cohort. Thirty-two single nucleotide polymorphisms (SNPs) in 21 genes across the EDC were genotyped in 402 unrelated AD patients and 325 non-atopic controls by means of restriction enzyme digestion or TaqMan assays. Allele and genotype frequencies were tested for differences between patients and controls by logistic regression. Haplotype frequencies were evaluated using the famhap software. Except for the already known association with FLG, we did not identify any additional significant associations of EDC genes with AD. Thus, in this German cohort, there is no evidence that additional genes in the EDC region apart from FLG contribute substantially to AD pathogenesis.
Assuntos
Diferenciação Celular/genética , Dermatite Atópica/genética , Epiderme/imunologia , Frequência do Gene/genética , Proteínas de Filamentos Intermediários/genética , Adolescente , Adulto , Alelos , Estudos de Casos e Controles , Criança , Pré-Escolar , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 1/imunologia , Dermatite Atópica/epidemiologia , Epiderme/patologia , Proteínas Filagrinas , Frequência do Gene/imunologia , Predisposição Genética para Doença/epidemiologia , Predisposição Genética para Doença/genética , Genótipo , Alemanha/epidemiologia , Haplótipos/genética , Haplótipos/imunologia , Humanos , Lactente , Proteínas de Filamentos Intermediários/imunologia , Desequilíbrio de Ligação/genética , Desequilíbrio de Ligação/imunologia , Polimorfismo de Nucleotídeo Único , Adulto JovemRESUMO
Multiple myeloma is incurable by standard approaches because of inevitable relapse and development of treatment resistance in all patients. In our prior work, we identified a panel of macropinocytosing human monoclonal antibodies against CD46, a negative regulator of the innate immune system, and constructed antibody-drug conjugates (ADCs). In this report, we show that an anti-CD46 ADC (CD46-ADC) potently inhibited proliferation in myeloma cell lines with little effect on normal cells. CD46-ADC also potently eliminated myeloma growth in orthometastatic xenograft models. In primary myeloma cells derived from bone marrow aspirates, CD46-ADC induced apoptosis and cell death, but did not affect the viability of nontumor mononuclear cells. It is of clinical interest that the CD46 gene resides on chromosome 1q, which undergoes genomic amplification in the majority of relapsed myeloma patients. We found that the cell surface expression level of CD46 was markedly higher in patient myeloma cells with 1q gain than in those with normal 1q copy number. Thus, genomic amplification of CD46 may serve as a surrogate for target amplification that could allow patient stratification for tailored CD46-targeted therapy. Overall, these findings indicate that CD46 is a promising target for antibody-based treatment of multiple myeloma, especially in patients with gain of chromosome 1q.
Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Antineoplásicos/farmacologia , Imunoconjugados/farmacologia , Proteína Cofatora de Membrana/antagonistas & inibidores , Mieloma Múltiplo/tratamento farmacológico , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Antineoplásicos/imunologia , Linhagem Celular Tumoral , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 1/imunologia , Dosagem de Genes/imunologia , Humanos , Imunoconjugados/imunologia , Proteína Cofatora de Membrana/genética , Proteína Cofatora de Membrana/imunologia , Camundongos , Mieloma Múltiplo/genética , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia , Ensaios Antitumorais Modelo de XenoenxertoRESUMO
Interleukin (IL)-20 belongs to the IL-10 family and is a potent immunomodulatory cytokine with implications for pathogenesis in the inflammatory bowel disease (IBD). The interleukin 20 gene is located within a 200kb region of q31-32 locus of chromosome 1. No previous studies have reported this novel association between ulcerative colitis (UC) and IL-20 polymorphisms. In the present work, we evaluated the role of IL-20 gene polymorphisms as susceptibility markers for UC. Three polymorphisms of IL-20 gene (rs2981573, rs2232360, rs1518108) were genotyped by 5' exonuclease TaqMan genotyping assays on an ABI Prism 7900 HT Fast Real-Time PCR system in a group of 198 Mexican Mestizo patients with UC and 698 ethnically matched healthy unrelated individuals with no family history of UC. We found significant decreased frequencies of two IL-20 genotypes: GG (rs2981573) [10.6% vs. 17.6%, p=0.017, OR=0.55, 95% CI: 0.33-0.93] and GG (rs2232360) [10.6% vs. 17.6%, p=0.017, OR=0.55, 95% CI: 0.33-0.93] in UC patients as compared to healthy controls. No significant differences of gene frequencies were found between UC patients and healthy controls in the rs1518108 polymorphism. In the subgroup analysis, no differences were found between the IL-20 genotypes and the clinical characteristics of UC. The results suggest that the GG genotypes of the IL-20 polymorphisms (rs2981573 and rs2232360) might have an important role in the development of UC in the Mexican population.
Assuntos
Cromossomos Humanos Par 1/genética , Colite Ulcerativa/genética , Predisposição Genética para Doença , Interleucinas/genética , Polimorfismo Genético , Adulto , Cromossomos Humanos Par 1/imunologia , Colite Ulcerativa/imunologia , Feminino , Humanos , Interleucinas/imunologia , Masculino , México , Pessoa de Meia-IdadeRESUMO
OBJECTIVE: Systemic lupus erythematosus is a multifactorial disease with a strong genetic component. Previous studies have shown that a 129-derived chromosome 1 interval (Sle16) on the C57BL/6 (B6) background is sufficient to induce humoral autoimmunity. The aim of the present study was to elucidate the mechanisms by which this locus contributes to the loss of peripheral tolerance. METHODS: Anti-single-stranded DNA (anti-ssDNA)-knockin transgenic mice (V(H)3H9R/Vkappa8R and V(H)3H9R) were crossed with a B6 congenic line named B6.129chr1b that carries the Sle16 locus. A parallel study of a gene-targeted animal, whose mutated gene is located within the 129chr1b interval on chromosome 1, was also performed. RESULTS: The combination of V(H)3H9R/Vkappa8R with the 129chr1b interval resulted in impaired B cell anergy, and transgenic IgM and IgG anti-ssDNA antibodies were found in the circulation. The presence of IgG2a(a) anti-ssDNA and IgM(a) anti-Sm antibodies in sera indicated that the autoreactive transgenic B cells underwent class switching and epitope spreading. The 129chr1b locus appeared to have a dominant effect, since transgenic antibodies were also detected in mice carrying a single allele. The gene-targeted animals showed a similar phenotype. CONCLUSION: The presence of a single 129chr1b locus on the B6 background impaired B cell anergy, prevented deletion of anti-DNA transgenic B cells, and induced receptor revision. The findings of this study also emphasize that the autoimmune phenotype observed in mice with targeted genes located on chromosome 1 may simply arise from epistatic interactions between the 129 and B6 parental strains.
Assuntos
Autoimunidade/imunologia , Linfócitos B/imunologia , Cromossomos Humanos Par 1/imunologia , Switching de Imunoglobulina/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Animais , Autoanticorpos , DNA de Cadeia Simples , Humanos , Imunoglobulina G , Imunoglobulina M , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , FenótipoRESUMO
Discovery of a large family of Fc receptor-like (FCRL) molecules, homologous to the well-known receptors for the Fc portion of immunoglobulin (FCR), has uncovered an impressive abundance of immunoglobulin superfamily (IgSF) genes in the human 1q21-23 chromosomal region and revealed significant diversity for these genes between humans and mice. The observation that FCRL representatives are members of an ancient multigene family that share a common ancestor with the classical FCR is underscored by their linked genomic locations, gene structure, shared extracellular domain composition, and utilization of common cytoplasmic tyrosine-based signaling elements. In contrast to the conventional FCR, however, FCRL molecules possess diverse extracellular frameworks, autonomous or dual signaling properties, and preferential B lineage expression. Most importantly, there is no strong evidence thus far to support a role for them as Ig-binding receptors. These characteristics, in addition to their identification in malignancies and autoimmune disorders, predict a fundamental role for these receptors as immunomodulatory agents in normal and subverted B lineage cells.
Assuntos
Linfócitos B/imunologia , Cromossomos Humanos Par 1/imunologia , Família Multigênica/imunologia , Receptores Imunológicos/imunologia , Transdução de Sinais/imunologia , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Cromossomos Humanos Par 1/genética , Regulação da Expressão Gênica/imunologia , Humanos , Família Multigênica/genética , Neoplasias/genética , Neoplasias/imunologia , Receptores Imunológicos/genética , Transdução de Sinais/genéticaRESUMO
The structural genes of human complement regulatory proteins are clustered on chromosome 1 at position q3.2. Human chromosome 1 was transferred into a mouse fibroblast cell line, A9 [designated as A9(neo-1)], and the surface expression of its gene products participating in complement regulation, namely C3b/C4b receptor (CR1, CD35), decay-accelerating factor (DAF, CD55), membrane co-factor protein (MCP, CD46) and C3d/EB virus receptor (CR2, CD21), were assessed using respective monoclonal antibodies by flow cytometry. CR1 became positive within 7 days of culture. MCP appeared in a small population of cells by Day 3 and, together with DAF, began to increase on Day 7. CR2 appeared on Day 14. The order of the expression was CR1 greater than DAF = MCP greater than CR2. On Day 42, however, all became negative except for MCP, which was markedly diminished. These human regulatory proteins were specifically associated with the presence of human chromosome 1, since none of them were expressed on human chromosome 12-transferred A9 cells [A9(neo-12)]. Intact A9 and A9(neo-12) cells activated human complement via the alternative pathway. The activation of this pathway was suppressed in the A9(neo-1) cells that expressed CR1, DAF and MCP. Slight protective activity was still observed in the 42-day cultured A9(neo-1) cells expressing only trace MCP. These results suggest that human complement regulators, expressed via the transferred human chromosome 1, can protect heterologous cells from complement, overcoming their ability to activate the human alternative pathway.
Assuntos
Cromossomos Humanos Par 1/imunologia , Proteínas Inativadoras do Complemento/genética , Via Alternativa do Complemento/genética , Animais , Antígenos CD/genética , Antígenos de Diferenciação de Linfócitos B/genética , Antígenos CD55 , Células Cultivadas , Epitopos/análise , Humanos , Proteína Cofatora de Membrana , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Proteínas de Membrana/genética , Camundongos , Receptores de Complemento/genética , Receptores de Complemento 3b , Receptores de Complemento 3dRESUMO
NKT cells play a critical role in shaping the character and strength of a wide range of immune responses, including those against pathogens, tumours, allografts and autologous tissues. Because numbers of NKT cells affect clinical outcomes in a wide range of disease models, and this characteristic demonstrates allelic variation, the mapping of the locations and identification of the coding sequences of these genes has become a matter of significant importance. Here, we review the results to date that examine the effects of targeted deletion of a number of candidate genes, as well as the congenic and genetic linkage analyses that have attempted to localize allelic loci that affect NKT cell numbers. Although a number of candidate genes have been examined, there is no evidence that any of these contribute to variation in NKT cell numbers in natural populations. Two of the most important genetic regions controlling NKT cell numbers are Nkt1 on chromosome 1, which may contribute to lupus susceptibility, and Nkt2 on chromosome 2, which appears to contribute to diabetes susceptibility. Of great interest is a third locus on chromosome 18, identified in a novel congenic line, which can confer an absolute deficiency in this important immunoregulatory lymphocyte population.
Assuntos
Cromossomos Humanos Par 18/genética , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 2/genética , Predisposição Genética para Doença , Células Matadoras Naturais , Ativação Linfocitária/genética , Linfócitos T , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Contagem de Células , Cromossomos Humanos Par 1/imunologia , Cromossomos Humanos Par 18/imunologia , Cromossomos Humanos Par 2/imunologia , Doenças Transmissíveis/genética , Doenças Transmissíveis/imunologia , Humanos , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Neoplasias/genética , Neoplasias/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Transplante HomólogoRESUMO
El estudio citogenético en la médula ósea de 61 pacientes pediátricos con hemopatías malignas mostró que 36 (59 por ciento) tenían un cariotopo anormal al momento del diagnóstico. De éstos, 15 tenían anormalidades estructurales y en ocho el cromosoma 1 estaba involucrado en alguno de los rearreglos. El objetivo del presente etudio fue analizar las alteraciones del cromosoma 1 y el estado clínico de los pacientes porque otros autores han observado la aparición de estos rearreglos en enfermos con recaída o en estado terminal. En tres casos la alteración en la que participaba el cromosoma 1 fueron primarias; y en los restantes fueron cambios clonales secundarios que aparecen durante la evolución de la enfermedad. Al momento del diagnóstico, cinco de estos pacientes tenían cuentas elevadas de leucocitos o enfermedad extramedular y la respuesta al tratamiento en general fue mala. Se asume que la alta frecuencia de alteraciones del cromosoma 1 en la población estudiada se debe a que los pacientes llegan al Instituto para su diagnóstico y tratamiento cuando su padecimiento está avanzado, existe evolución clonal de las células leucémicas y el pronóstico es malo.