Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 14: 1145269, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37600780

RESUMO

Leishmaniasis, a disease caused by parasites of Leishmania spp., endangers more than 1 billion people living in endemic countries and has three clinical forms: cutaneous, mucocutaneous, and visceral. Understanding of individual differences in susceptibility to infection and heterogeneity of its pathology is largely lacking. Different mouse strains show a broad and heterogeneous range of disease manifestations such as skin lesions, splenomegaly, hepatomegaly, and increased serum levels of immunoglobulin E and several cytokines. Genome-wide mapping of these strain differences detected more than 30 quantitative trait loci (QTLs) that control the response to Leishmania major. Some control different combinations of disease manifestations, but the nature of this heterogeneity is not yet clear. In this study, we analyzed the L. major response locus Lmr15 originally mapped in the strain CcS-9 which carries 12.5% of the genome of the resistant strain STS on the genetic background of the susceptible strain BALB/c. For this analysis, we used the advanced intercross line K3FV between the strains BALB/c and STS. We confirmed the previously detected loci Lmr15, Lmr18, Lmr24, and Lmr27 and performed genetic dissection of the effects of Lmr15 on chromosome 11. We prepared the interval-specific recombinant strains 6232HS1 and 6229FUD, carrying two STS-derived segments comprising the peak linkage of Lmr15 whose lengths were 6.32 and 17.4 Mbp, respectively, and analyzed their response to L. major infection. These experiments revealed at least two linked but functionally distinct chromosomal regions controlling IFNγ response and IgE response, respectively, in addition to the control of skin lesions. Bioinformatics and expression analysis identified the potential candidate gene Top3a. This finding further clarifies the genetic organization of factors relevant to understanding the differences in the individual risk of disease.


Assuntos
Leishmania major , Dermatopatias , Animais , Camundongos , Leishmania major/genética , Interferon gama/genética , Citocinas , Imunoglobulina E
2.
Front Immunol ; 12: 760881, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35154069

RESUMO

Differences in frequencies of blood cell subpopulations were reported to influence the course of infections, atopic and autoimmune diseases, and cancer. We have discovered a unique mouse strain B10.O20 containing extremely high frequency of myeloid-derived cells (MDC) in spleen. B10.O20 carries 3.6% of genes of the strain O20 on the C57BL/10 genetic background. It contains much higher frequency of CD11b+Gr1+ cells in spleen than both its parents. B10.O20 carries O20-derived segments on chromosomes 1, 15, 17, and 18. Their linkage with frequencies of blood cell subpopulations in spleen was tested in F2 hybrids between B10.O20 and C57BL/10. We found 3 novel loci controlling MDC frequencies: Mydc1, 2, and 3 on chromosomes 1, 15, and 17, respectively, and a locus controlling relative spleen weight (Rsw1) that co-localizes with Mydc3 and also influences proportion of white and red pulp in spleen. Mydc1 controls numbers of CD11b+Gr1+ cells. Interaction of Mydc2 and Mydc3 regulates frequency of CD11b+Gr1+ cells and neutrophils (Gr1+Siglec-F- cells from CD11b+ cells). Interestingly, Mydc3/Rsw1 is orthologous with human segment 6q21 that was shown previously to determine counts of white blood cells. Bioinformatics analysis of genomic sequence of the chromosomal segments bearing these loci revealed polymorphisms between O20 and C57BL/10 that change RNA stability and genes' functions, and we examined expression of relevant genes. This identified potential candidate genes Smap1, Vps52, Tnxb, and Rab44. Definition of genetic control of MDC can help to personalize therapy of diseases influenced by these cells.


Assuntos
Células Mieloides/fisiologia , Animais , Cromossomos/genética , Biologia Computacional/métodos , Feminino , Ligação Genética/genética , Loci Gênicos/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/fisiologia , Polimorfismo Genético/genética , Estabilidade de RNA/genética , Baço/fisiologia
3.
Front Immunol ; 10: 1083, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31231359

RESUMO

Leishmaniasis is a serious health problem in many countries, and continues expanding to new geographic areas including Europe and USA. This disease, caused by parasites of Leishmania spp. and transmitted by phlebotomine sand flies, causes up to 1.3 million new cases each year and despite efforts toward its functional dissection and treatment it causes 20-50 thousands deaths annually. Dependence of susceptibility to leishmaniasis on sex and host's genes was observed in humans and in mouse models. Several laboratories defined in mice a number of Lmr (Leishmania major response) genetic loci that control functional and pathological components of the response to and outcome of L. major infection. However, the development of its most aggressive form, visceral leishmaniasis, which is lethal if untreated, is not yet understood. Visceral leishmaniasis is caused by infection and inflammation of internal organs. Therefore, we analyzed the genetics of parasite load, spread to internal organs, and ensuing visceral pathology. Using a new PCR-based method of quantification of parasites in tissues we describe a network-like set of interacting genetic loci that control parasite load in different organs. Quantification of Leishmania parasites in lymph nodes, spleen and liver from infected F2 hybrids between BALB/c and recombinant congenic strains CcS-9 and CcS-16 allowed us to map two novel parasite load controlling Leishmania major response loci, Lmr24 and Lmr27. We also detected parasite-controlling role of the previously described loci Lmr4, Lmr11, Lmr13, Lmr14, Lmr15, and Lmr25, and describe 8 genetic interactions between them. Lmr14, Lmr15, Lmr25, and Lmr27 controlled parasite load in liver and lymph nodes. In addition, Leishmania burden in lymph nodes but not liver was influenced by Lmr4 and Lmr24. In spleen, parasite load was controlled by Lmr11 and Lmr13. We detected a strong effect of sex on some of these genes. We also mapped additional genes controlling splenomegaly and hepatomegaly. This resulted in a systematized insight into genetic control of spread and load of Leishmania parasites and visceral pathology in the mammalian organism.


Assuntos
Leishmania major , Leishmaniose Visceral/genética , Leishmaniose Visceral/parasitologia , Carga Parasitária , Animais , Feminino , Interações Hospedeiro-Parasita , Masculino , Camundongos , Caracteres Sexuais
4.
Front Immunol ; 9: 130, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29467757

RESUMO

Interferon-induced GTPases [guanylate-binding proteins (GBPs)] play an important role in inflammasome activation and mediate innate resistance to many intracellular pathogens, but little is known about their role in leishmaniasis. We therefore studied expression of Gbp2b/Gbp1 and Gbp5 mRNA in skin, inguinal lymph nodes, spleen, and liver after Leishmania major infection and in uninfected controls. We used two different groups of related mouse strains: BALB/c, STS, and CcS-5, CcS-16, and CcS-20 that carry different combinations of BALB/c and STS genomes, and strains O20, C57BL/10 (B10) and B10.O20, OcB-9, and OcB-43 carrying different combinations of O20 and B10 genomes. The strains were classified on the basis of size and number of infection-induced skin lesions as highly susceptible (BALB/c, CcS-16), susceptible (B10.O20), intermediate (CcS-20), and resistant (STS, O20, B10, OcB-9, OcB-43). Some uninfected strains differed in expression of Gbp2b/Gbp1 and Gbp5, especially of Gbp2b/Gbp1 in skin. Uninfected BALB/c and STS did not differ in their expression, but in CcS-5, CcS-16, and CcS-20, which all carry BALB/c-derived Gbp gene-cluster, expression of Gbp2b/Gbp1 exceeds that of both parents. These data indicate trans-regulation of Gbps. Infection resulted in approximately 10× upregulation of Gbp2b/Gbp1 and Gbp5 mRNAs in organs of both susceptible and resistant strains, which was most pronounced in skin. CcS-20 expressed higher level of Gbp2b/Gbp1 than both parental strains in skin, whereas CcS-16 expressed higher level of Gbp2b/Gbp1 than both parental strains in skin and liver. This indicates a trans-regulation present in infected mice CcS-16 and CcS-20. Immunostaining of skin of five strains revealed in resistant and intermediate strains STS, CcS-5, O20, and CcS-20 tight co-localization of Gbp2b/Gbp1 protein with most L. major parasites, whereas in the highly susceptible strain, BALB/c most parasites did not associate with Gbp2b/Gbp1. In conclusion, expression of Gbp2b/Gbp1 and Gbp5 was increased even in organs of clinically asymptomatic resistant mice. It suggests a hidden inflammation, which might contribute to control of persisting parasites. This is supported by the co-localization of Gbpb2/Gbp1 protein and L. major parasites in skin of resistant and intermediate but not highly susceptible mice.


Assuntos
Proteínas de Ligação ao GTP/genética , Leishmaniose Cutânea/genética , Animais , Feminino , Regulação da Expressão Gênica , Fígado/metabolismo , Linfonodos/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Pele/metabolismo , Especificidade da Espécie , Baço/metabolismo
5.
J Hepatol ; 68(5): 1006-1017, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29273475

RESUMO

BACKGROUND & AIMS: Plectin, a highly versatile cytolinker protein, controls intermediate filament cytoarchitecture and cellular stress response. In the present study, we investigate the role of plectin in the liver under basal conditions and in experimental cholestasis. METHODS: We generated liver-specific plectin knockout (PleΔalb) mice and analyzed them using two cholestatic liver injury models: bile duct ligation (BDL) and 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) feeding. Primary hepatocytes and a cholangiocyte cell line were used to address the impact of plectin on keratin filament organization and stability in vitro. RESULTS: Plectin deficiency in hepatocytes and biliary epithelial cells led to aberrant keratin filament network organization, biliary tree malformations, and collapse of bile ducts and ductules. Further, plectin ablation significantly aggravated biliary damage upon cholestatic challenge. Coincidently, we observed a significant expansion of A6-positive progenitor cells in PleΔalb livers. After BDL, plectin-deficient bile ducts were prominently dilated with more frequent ruptures corresponding to an increased number of bile infarcts. In addition, more abundant keratin aggregates indicated less stable keratin filaments in PleΔalb hepatocytes. A transmission electron microscopy analysis revealed a compromised tight junction formation in plectin-deficient biliary epithelial cells. In addition, protein profiling showed increased expression of the adherens junction protein E-Cadherin, and inefficient upregulation of the desmosomal protein desmoplakin in response to BDL. In vitro analyses revealed a higher susceptibility of plectin-deficient keratin networks to stress-induced collapse, paralleled by elevated activation of p38 MAP kinase. CONCLUSION: Our study shows that by maintaining proper keratin network cytoarchitecture and biliary epithelial stability, plectin plays a critical role in protecting the liver from stress elicited by cholestasis. LAY SUMMARY: Plectin is a cytolinker protein capable of interconnecting all three cytoskeletal filament systems and linking them to plasma membrane-bound junctional complexes. In liver, the plectin-controlled cytoskeleton mechanically stabilizes epithelial cells and provides them with the capacity to adapt to increased bile pressure under cholestasis.


Assuntos
Sistema Biliar/metabolismo , Sistema Biliar/patologia , Colestase/metabolismo , Colestase/patologia , Plectina/metabolismo , Animais , Sistema Biliar/anormalidades , Epitélio/metabolismo , Epitélio/patologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Queratinas/metabolismo , Fígado/anormalidades , Fígado/metabolismo , Fígado/patologia , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Knockout , Plectina/deficiência , Plectina/genética , Estabilidade Proteica , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
PLoS Negl Trop Dis ; 7(7): e2282, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23875032

RESUMO

BACKGROUND: L. tropica can cause both cutaneous and visceral leishmaniasis in humans. Although the L. tropica-induced cutaneous disease has been long known, its potential to visceralize in humans was recognized only recently. As nothing is known about the genetics of host responses to this infection and their clinical impact, we developed an informative animal model. We described previously that the recombinant congenic strain CcS-16 carrying 12.5% genes from the resistant parental strain STS/A and 87.5% genes from the susceptible strain BALB/c is more susceptible to L. tropica than BALB/c. We used these strains to map and functionally characterize the gene-loci regulating the immune responses and pathology. METHODS: We analyzed genetics of response to L. tropica in infected F2 hybrids between BALB/c×CcS-16. CcS-16 strain carries STS-derived segments on nine chromosomes. We genotyped these segments in the F2 hybrid mice and tested their linkage with pathological changes and systemic immune responses. PRINCIPAL FINDINGS: We mapped 8 Ltr (Leishmania tropica response) loci. Four loci (Ltr2, Ltr3, Ltr6 and Ltr8) exhibit independent responses to L. tropica, while Ltr1, Ltr4, Ltr5 and Ltr7 were detected only in gene-gene interactions with other Ltr loci. Ltr3 exhibits the recently discovered phenomenon of transgenerational parental effect on parasite numbers in spleen. The most precise mapping (4.07 Mb) was achieved for Ltr1 (chr.2), which controls parasite numbers in lymph nodes. Five Ltr loci co-localize with loci controlling susceptibility to L. major, three are likely L. tropica specific. Individual Ltr loci affect different subsets of responses, exhibit organ specific effects and a separate control of parasite load and organ pathology. CONCLUSION: We present the first identification of genetic loci controlling susceptibility to L. tropica. The different combinations of alleles controlling various symptoms of the disease likely co-determine different manifestations of disease induced by the same pathogen in individual mice.


Assuntos
Mapeamento Cromossômico , Suscetibilidade a Doenças , Interações Hospedeiro-Patógeno , Leishmaniose Cutânea/genética , Animais , Modelos Animais de Doenças , Feminino , Loci Gênicos , Camundongos
7.
PLoS Negl Trop Dis ; 6(6): e1667, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22679519

RESUMO

BACKGROUND: Leishmaniasis is a disease caused by protozoan parasites of genus Leishmania. The frequent involvement of Leishmania tropica in human leishmaniasis has been recognized only recently. Similarly as L. major, L. tropica causes cutaneous leishmaniasis in humans, but can also visceralize and cause systemic illness. The relationship between the host genotype and disease manifestations is poorly understood because there were no suitable animal models. METHODS: We studied susceptibility to L. tropica, using BALB/c-c-STS/A (CcS/Dem) recombinant congenic (RC) strains, which differ greatly in susceptibility to L. major. Mice were infected with L. tropica and skin lesions, cytokine and chemokine levels in serum, and parasite numbers in organs were measured. PRINCIPAL FINDINGS: Females of BALB/c and several RC strains developed skin lesions. In some strains parasites visceralized and were detected in spleen and liver. Importantly, the strain distribution pattern of symptoms caused by L. tropica was different from that observed after L. major infection. Moreover, sex differently influenced infection with L. tropica and L. major. L. major-infected males exhibited either higher or similar skin pathology as females, whereas L. tropica-infected females were more susceptible than males. The majority of L. tropica-infected strains exhibited increased levels of chemokines CCL2, CCL3 and CCL5. CcS-16 females, which developed the largest lesions, exhibited a unique systemic chemokine reaction, characterized by additional transient early peaks of CCL3 and CCL5, which were not present in CcS-16 males nor in any other strain. CONCLUSION: Comparison of L. tropica and L. major infections indicates that the strain patterns of response are species-specific, with different sex effects and largely different host susceptibility genes.


Assuntos
Suscetibilidade a Doenças , Interações Hospedeiro-Patógeno , Leishmania major/imunologia , Leishmania tropica/imunologia , Leishmaniose Cutânea/genética , Leishmaniose Cutânea/parasitologia , Animais , Citocinas/sangue , Modelos Animais de Doenças , Feminino , Humanos , Leishmania major/patogenicidade , Leishmania tropica/patogenicidade , Leishmaniose Cutânea/imunologia , Fígado/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Carga Parasitária , Fatores Sexuais , Pele/parasitologia , Pele/patologia , Baço/parasitologia
8.
PLoS Negl Trop Dis ; 5(6): e1173, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21666791

RESUMO

BACKGROUND: Trypanosoma brucei brucei infects livestock, with severe effects in horses and dogs. Mouse strains differ greatly in susceptibility to this parasite. However, no genes controlling these differences were mapped. METHODS: We studied the genetic control of survival after T. b. brucei infection using recombinant congenic (RC) strains, which have a high mapping power. Each RC strain of BALB/c-c-STS/A (CcS/Dem) series contains a different random subset of 12.5% genes from the parental "donor" strain STS/A and 87.5% genes from the "background" strain BALB/c. Although BALB/c and STS/A mice are similarly susceptible to T. b. brucei, the RC strain CcS-11 is more susceptible than either of them. We analyzed genetics of survival in T. b. brucei-infected F(2) hybrids between BALB/c and CcS-11. CcS-11 strain carries STS-derived segments on eight chromosomes. They were genotyped in the F(2) hybrid mice and their linkage with survival was tested by analysis of variance. RESULTS: We mapped four Tbbr (Trypanosoma brucei brucei response) loci that influence survival after T. b. brucei infection. Tbbr1 (chromosome 3) and Tbbr2 (chromosome 12) have effects on survival independent of inter-genic interactions (main effects). Tbbr3 (chromosome 7) influences survival in interaction with Tbbr4 (chromosome 19). Tbbr2 is located on a segment 2.15 Mb short that contains only 26 genes. CONCLUSION: This study presents the first identification of chromosomal loci controlling susceptibility to T. b. brucei infection. While mapping in F(2) hybrids of inbred strains usually has a precision of 40-80 Mb, in RC strains we mapped Tbbr2 to a 2.15 Mb segment containing only 26 genes, which will enable an effective search for the candidate gene. Definition of susceptibility genes will improve the understanding of pathways and genetic diversity underlying the disease and may result in new strategies to overcome the active subversion of the immune system by T. b. brucei.


Assuntos
Imunidade Inata/genética , Doenças dos Roedores/genética , Doenças dos Roedores/imunologia , Trypanosoma brucei brucei/imunologia , Trypanosoma brucei brucei/patogenicidade , Tripanossomíase Africana/genética , Tripanossomíase Africana/imunologia , Animais , Mapeamento Cromossômico , Cruzamentos Genéticos , Feminino , Loci Gênicos , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sobrevida
9.
Nat Protoc ; 5(6): 1074-80, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20539283

RESUMO

This protocol describes an improved and optimized PCR-ELISA method for detection and quantification of Leishmania parasites in host tissues. Unlike other DNA-based assays, this method uses digoxigenin- and biotin-labeled primers. This eliminates the need for a separate step of hybridization of the PCR product with labeled probes. The PCR product is detected using sandwich ELISA with antidigoxigenin-detecting antibodies. Primers are complementary to the kinetoplast minicircle conserved region of parasite DNA, allowing the detection of several Leishmania species. For measurement of a wide range of parasite concentrations, +/-25 cycles were optimal. The sensitivity of this technique is 0.3 fg of parasite DNA per reaction in 40-cycle PCR-ELISA, corresponding to 0.004 parasites. DNA preparation by a standard TRI reagent procedure takes about 4 h. When DNA is prepared, a single person can test a large number of samples (at least 150) in a maximum of 7 h. This method might also be suitable for detecting and quantifying other pathogens, especially for detecting small differences in pathogen numbers.


Assuntos
Ensaio de Imunoadsorção Enzimática/métodos , Leishmania/genética , Leishmania/isolamento & purificação , Reação em Cadeia da Polimerase/métodos , Animais , Sequência de Bases , Primers do DNA/genética , DNA de Protozoário/análise , DNA de Protozoário/genética , Leishmania major/genética , Leishmania major/isolamento & purificação , Leishmaniose/diagnóstico , Leishmaniose/parasitologia , Camundongos
10.
Cancer Immunol Immunother ; 59(2): 203-13, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19655140

RESUMO

Low infiltration of lymphocytes into cancers is associated with poor prognosis, but the reasons why some patients exhibit a low and others a high infiltration of tumors are unknown. Previously we mapped four loci (Lynf1­Lynf4) controlling lymphocyte infiltration of mouse lung tumors. These loci do not encode any of the molecules that are involved in traffic of lymphocytes. Here we report a genetic relationship between these loci and the control of production of IFNγ in allogeneic mixed lymphocyte cultures (MLC). We found that IFNγ production by lymphocytes of O20/A mice is lower than by lymphocytes of OcB-9/Dem mice (both H2pz) stimulated in MLC by irradiated splenocytes of C57BL/10SnPh (H2b) or BALB/ cHeA (H2d) mice, or by ConA. IFNγ production in MLCs of individual (O20 9 OcB-9)F2mice stimulated by irradiated C57BL/10 splenocytes and genotyped for microsatellite markers revealed four IFNγ-controlling loci (Cypr4-Cypr7), each of which is closely linked with one of the four Lynf loci and with a cluster of susceptibility genes for different tumors. This suggests that inherited differences in certain lymphocyte responses may modify their propensity to infiltrate tumors and their capacity to affect tumor growth.


Assuntos
Loci Gênicos , Interferon gama/biossíntese , Isoantígenos/imunologia , Linfócitos do Interstício Tumoral/imunologia , Animais , Mapeamento Cromossômico , Feminino , Teste de Cultura Mista de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Mutantes
11.
Immunogenetics ; 61(9): 619-33, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19705113

RESUMO

Elimination of pathogens is the basis of host resistance to infections; however, relationship between persisting pathogens and disease has not been clarified. Leishmania major infection in mice is an important model of host-pathogen relationship. Infected BALB/c mice exhibit high parasite numbers in lymph nodes and spleens, and a chronic disease with skin lesions, splenomegaly, and hepatomegaly, increased serum IgE levels and cytokine imbalance. Although numerous gene loci affecting these disease symptoms have been reported, genes controlling parasites' elimination or dissemination have never been mapped. We therefore compared genetics of the clinical and immunologic symptomatology with parasite load in (BALB/c x CcS-11) F2 hybrids and mapped five loci, two of which control parasite elimination or dissemination. Lmr5 influences parasite loads in spleens (and skin lesions, splenomegaly, and serum IgE, IL-4, and IFNgamma levels), and Lmr20 determines parasite numbers in draining lymph nodes (and serum levels of IgE and IFNgamma), but no skin or visceral pathology. Three additional loci do not affect parasite numbers but influence significantly the disease phenotype-Lmr21: skin lesions and IFNgamma levels, Lmr22: IL-4 levels, Lmr23: IFNgamma levels, indicating that development of L. major-caused disease includes critical regulations additional to control of parasite spread.


Assuntos
Leishmania major/imunologia , Leishmaniose Cutânea/genética , Leishmaniose Cutânea/imunologia , Animais , Feminino , Interferon gama/sangue , Leishmaniose Cutânea/parasitologia , Leishmaniose Cutânea/patologia , Linfonodos/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Caracteres Sexuais , Pele/patologia , Baço/parasitologia
12.
Immunogenetics ; 61(1): 15-25, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19015841

RESUMO

Atopy is a predisposition to hyperproduction of immunoglobulin E (IgE) against common environmental allergens. It is often associated with development of allergic diseases such as asthma, rhinitis, and dermatitis. Production of IgE is influenced by genetic and environmental factors. In spite of progress in the study of heredity of atopy, the genetic mechanisms of IgE regulation have not yet been completely elucidated. The analysis of complex traits can benefit considerably from integration of human and mouse genetics. Previously, we mapped a mouse IgE-controlling locus Lmr9 on chromosome 4 to a segment of <9 Mb. In this study, we tested levels of total IgE and 25 specific IgEs against inhalant and food allergens in 67 Czech atopic families. In the position homologous to Lmr9 on chromosome 8q12 marked by D8S285, we demonstrated a novel human IgE-controlling locus exhibiting suggestive linkage to composite inhalant allergic sensitization (limit of detection, LOD = 2.11, P = 0.0009) and to nine specific IgEs, with maximum LOD (LOD = 2.42, P = 0.0004) to plantain. We also tested 16 markers at previously reported chromosomal regions of atopy. Linkage to plant allergens exceeding the LOD > 2.0 was detected at 5q33 (D5S1507, LOD = 2.11, P = 0.0009) and 13q14 (D13S165, LOD = 2.74, P = 0.0002). The significant association with plant allergens (quantitative and discrete traits) was found at 7p14 (D7S2250, corrected P = 0.026) and 12q13 (D12S1298, corrected P = 0.043). Thus, the finding of linkage on chromosome 8q12 shows precision and predictive power of mouse models in the investigation of complex traits in humans. Our results also confirm the role of loci at 5q33, 7p14, 12q14, and 13q13 in control of IgE.


Assuntos
Alérgenos/imunologia , Mapeamento Cromossômico , Cromossomos Humanos Par 8/genética , Epigênese Genética/genética , Hipersensibilidade Imediata/genética , Imunoglobulina E/biossíntese , Camundongos/genética , Adolescente , Adulto , Alérgenos/efeitos adversos , Animais , Criança , República Tcheca/epidemiologia , Dermatite Atópica/etnologia , Dermatite Atópica/genética , Dermatite Atópica/imunologia , Etnicidade/genética , Feminino , Hipersensibilidade Alimentar/etnologia , Hipersensibilidade Alimentar/genética , Hipersensibilidade Alimentar/imunologia , Predisposição Genética para Doença , Humanos , Hipersensibilidade Imediata/etnologia , Imunoglobulina E/sangue , Imunoglobulina E/imunologia , Escore Lod , Masculino , Camundongos/imunologia , Pessoa de Meia-Idade , Característica Quantitativa Herdável , Hipersensibilidade Respiratória/etnologia , Hipersensibilidade Respiratória/genética , Hipersensibilidade Respiratória/imunologia , Especificidade da Espécie
13.
Int J Cancer ; 114(3): 394-9, 2005 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-15551356

RESUMO

Novel genotyping and statistical tools have led to mapping of numerous QTL loci for multigenic traits that previously could not be detected. The relationships of these QTL families to other QTL families and the functional specialization of their members can now be studied. We have mapped a number of loci controlling activation of T lymphocytes by mitogens and cytokines and their capacity to produce cytokines. In (O20xOcB-9)F2 hybrids, we mapped 3 novel loci controlling proliferative T-cell response to cytokines IL-2 and IL-4 (Cinda3) or IL-4 only (Cinda4 and Cinda5). OcB-9 allele at Cinda3 controls a higher response than the O20 allele to both IL-2 and IL-4, and OcB-9 alleles of Cinda4 and Cinda5 control higher response to IL-4. These novel Cinda loci and the previously mapped Cinda1 locus seem to be located in genomic regions together with other QTL families: macrophage function loci Marif1 and Marif2, proteoglycan-induced arthritis loci Pgia4, Pgia7 and Pgia12 and lung tumor susceptibility loci Sluc1, Sluc4, Sluc6 and Sluc20. The possible relevance of these QTL associations in several different sites of the genome for the immune response, inflammation and tumorigenesis has to be elucidated.


Assuntos
Autoimunidade/imunologia , Inflamação , Neoplasias Pulmonares/etiologia , Neoplasias Pulmonares/imunologia , Linfócitos/fisiologia , Locos de Características Quantitativas , Animais , Proliferação de Células , Transformação Celular Neoplásica , Citocinas/farmacologia , Feminino , Predisposição Genética para Doença , Genoma , Genótipo , Neoplasias Pulmonares/genética , Macrófagos/fisiologia , Masculino , Camundongos
14.
Infect Immun ; 71(4): 2041-6, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12654824

RESUMO

The mouse strains BALB/cHeA (BALB/c) and STS/A (STS) are susceptible and resistant to Leishmania major-induced disease, respectively. We analyzed this difference using recombinant congenic (RC) BALB/c-c-STS/Dem (CcS/Dem) strains that carry different random subsets of 12.5% genes of the strain STS in a BALB/c background. Previously, testing the resistant strain CcS-5, we found five novel Lmr (Leishmania major response) loci, each associated with a different combination of pathological and immunological reactions. Here we analyze the response of RC strain CcS-16, which is even more susceptible to L. major than BALB/c. In the (CcS-16 x BALB/c)F(2) hybrids we mapped three novel loci that influence cutaneous or visceral pathology. Lmr14 (chromosome 2) controls splenomegaly and hepatomegaly. On the other hand Lmr15 (chromosome 11) determines hepatomegaly only, and Lmr13 (chromosome 18) determines skin lesions only. These data confirm the complex control of L. major-induced pathology, where cutaneous and visceral pathology are controlled by different combinations of genes. It indicates organ-specific control of antiparasite responses. The definition of genes controlling these responses will permit a better understanding of pathways and genetic diversity underlying the different disease phenotypes.


Assuntos
Predisposição Genética para Doença , Leishmania major/patogenicidade , Leishmaniose Cutânea/genética , Leishmaniose Cutânea/imunologia , Leishmaniose Visceral/genética , Leishmaniose Visceral/imunologia , Animais , Mapeamento Cromossômico , Cruzamentos Genéticos , Feminino , Ligação Genética , Genótipo , Hepatomegalia , Leishmaniose Cutânea/fisiopatologia , Leishmaniose Visceral/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C/genética , Camundongos Endogâmicos/genética , Esplenomegalia
15.
Immunogenetics ; 54(3): 174-83, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12073146

RESUMO

Systematic assessment of the role of host genes in clinico-pathological and immunological manifestations of Leishmania major-induced disease in mice was performed using 20 recombinant congenic (RC) strains. As the RC strains are homozygous and each carries a different, random set of 12.5% genes from the resistant strain, STS/A, and 87.5% genes from the susceptible strain, BALB/cHeA, they allowed us to study the pathological and immunological characteristics of infected hosts in 20 fixed different random combinations of BALB/c and STS genes. The 20 RC strains differ widely in expression of different symptoms of disease and in immunological characteristics. Disease or healing in different strains occurred in association with different components of immune response -- with the exception of a frequently occurring correlation between the disease and IgE levels. Moreover, some parameters of the immune response were highly correlated in some strains but not at all in others. This shows that several patterns of the immune response may be associated with the same clinical outcome, depending on the host genotype. Our data also suggest that despite the complexity of regulation, when a sufficient number of controlling loci is known, the prediction of a phenotype is possible. Combining functional and clinical information with multilocus genotyping may improve our ability to predict the progression of the disease and to optimize the treatment.


Assuntos
Modelos Animais de Doenças , Leishmaniose/genética , Leishmaniose/imunologia , Camundongos Congênicos/genética , Camundongos Congênicos/imunologia , Animais , Citocinas/sangue , Feminino , Predisposição Genética para Doença , Variação Genética , Imunoglobulina E/sangue , Cinética , Leishmania major , Leishmaniose/patologia , Camundongos , Camundongos Endogâmicos BALB C , Modelos Genéticos , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA