Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Biol Chem ; 402(9): 1147-1154, 2021 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-34087963

RESUMO

Lymphotoxin-ß-receptor deficient (LTßR-/-) and Tumor Necrosis Factor Receptor p55 deficient (TNFRp55-/-) mice show defects in liver regeneration (LR) after partial hepatectomy (PHx) with significantly increased mortality. LTßR and TNFRp55 belong to the core members of the TNF/TNFR superfamily. Interestingly, combined failure of LTßR and TNFRp55 signaling after PHx leads to a complete defect in LR. Here, we first addressed the question which liver cell population crucially requires LTßR signaling for efficient LR. To this end, mice with a conditionally targeted LTßR allele (LTßRfl/fl) were crossed to AlbuminCre and LysozymeMCre mouse lines to unravel the function of the LTßR on hepatocytes and monocytes/macrophages/Kupffer cells, respectively. Analysis of these mouse lines clearly reveals that LTßR is required on hepatocytes for efficient LR while no deficit in LR was found in LTßRfl/fl × LysMCre mice. Second, the molecular basis for the cooperating role of LTßR and TNFRp55 signaling pathways in LR was investigated by transcriptome analysis of etanercept treated LTßR-/- (LTßR-/-/ET) mice. Bioinformatic analysis and subsequent verification by qRT-PCR identified novel target genes (Cyclin-L2, Fas-Binding factor 1, interferon-related developmental regulator 1, Leucyl-tRNA Synthetase 2, and galectin-4) that are upregulated by LTßR/TNFRp55 signaling after PHx and fail to be upregulated after PHx in LTßR-/-/ET mice.


Assuntos
Regeneração Hepática , Animais , Hepatectomia , Hepatócitos , Linfotoxina-beta , Transdução de Sinais
2.
Infect Immun ; 89(6)2021 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-33753412

RESUMO

The lymphotoxin ß receptor (LTßR) plays an essential role in the initiation of immune responses to intracellular pathogens. In mice, the LTßR is crucial for surviving acute toxoplasmosis; however, until now, a functional analysis was largely incomplete. Here, we demonstrate that the LTßR is a key regulator required for the intricate balance of adaptive immune responses. Toxoplasma gondii-infected LTßR-deficient (LTßR-/-) mice show globally altered interferon-γ (IFN-γ) regulation, reduced IFN-γ-controlled host effector molecule expression, impaired T cell functionality, and an absent anti-parasite-specific IgG response, resulting in a severe loss of immune control of the parasites. Reconstitution of LTßR-/- mice with toxoplasma immune serum significantly prolongs survival following T. gondii infection. Notably, analysis of RNA-seq data clearly indicates a specific effect of T. gondii infection on the B cell response and isotype switching. This study uncovers the decisive role of the LTßR in cytokine regulation and adaptive immune responses to control T. gondii.


Assuntos
Imunidade Adaptativa , Interações Hospedeiro-Parasita/imunologia , Imunidade Inata , Receptor beta de Linfotoxina/metabolismo , Toxoplasma/imunologia , Toxoplasmose/imunologia , Toxoplasmose/metabolismo , Animais , Modelos Animais de Doenças , Receptor beta de Linfotoxina/genética , Camundongos , Camundongos Knockout , Toxoplasmose/parasitologia
3.
J Immunol ; 205(2): 323-328, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32540999

RESUMO

TNF blockade is a successful treatment for human autoimmune disorders like rheumatoid arthritis and inflammatory bowel disease yet increases susceptibility to tuberculosis and other infections. The C-type lectin receptors (CLR) MINCLE, MCL, and DECTIN-2 are expressed on myeloid cells and sense mycobacterial cell wall glycolipids. In this study, we show that TNF is sufficient to upregulate MINCLE, MCL, and DECTIN-2 in macrophages. TNF signaling through TNFR1 p55 was required for upregulation of these CLR and for cytokine secretion in macrophages stimulated with the MINCLE ligand trehalose-6,6-dibehenate or infected with Mycobacterium bovis bacillus Calmette-Guérin. The Th17 response to immunization with the MINCLE-dependent adjuvant trehalose-6,6-dibehenate was specifically abrogated in TNF-deficient mice and strongly attenuated by TNF blockade with etanercept. Together, interference with production or signaling of TNF antagonized the expression of DECTIN-2 family CLR, thwarting vaccine responses and possibly increasing infection risk.


Assuntos
Lectinas Tipo C/metabolismo , Mycobacterium bovis/fisiologia , Receptores Imunológicos/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Células Th17/imunologia , Tuberculose/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Células Cultivadas , Etanercepte/administração & dosagem , Lectinas Tipo C/genética , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Imunológicos/genética , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Fosfatos Açúcares/metabolismo , Trealose/análogos & derivados , Trealose/metabolismo , Tuberculose/veterinária , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/genética
4.
Artigo em Inglês | MEDLINE | ID: mdl-31231617

RESUMO

Toxoplasma gondii (T. gondii) is an obligate intracellular parasite and belongs to the phylum Apicomplexa. T. gondii is of medical and veterinary importance, because T. gondii causes the parasitic disease toxoplasmosis. In human cells, the interferon-gamma inducible indoleamine 2,3-dioxygenase 1 (IDO1) is an antimicrobial effector mechanism that degrades tryptophan to kynurenine and thus limits pathogen proliferation in vitro. Furthermore, IDO is described to have immunosuppressive properties, e.g., regulatory T cell differentiation and T cell suppression in humans and mice. However, there is only little known about the role of IDO1 in mice during acute toxoplasmosis. To shed further light on the role of mIDO1 in vivo, we have used a specifically adjusted experimental model. Therein, we infected mIDO1-deficient (IDO-/-) C57BL/6 mice and appropriate wild-type (WT) control mice with a high dose of T. gondii ME49 tachyozoites (type II strain) via the intraperitoneal route and compared the phenotype of IDO-/- and WT mice during acute toxoplasmosis. During murine T. gondii infection, we found mIDO1 mRNA and mIDO1 protein, as well as mIDO1-mediated tryptophan degradation in lungs of WT mice. IDO-/- mice show no tryptophan degradation in the lung during infection. Even though T. gondii is tryptophan auxotroph and rapidly replicates during acute infection, the parasite load was similar in IDO-/- mice compared to WT mice 7 days post-infection. IDO1 is described to have immunosuppressive properties, and since T cell suppression is observed during acute toxoplasmosis, we analyzed the possible involvement of mIDO1. Here, we did not find differences in the intensity of ex vivo mitogen stimulated T cell proliferation between WT and IDO-/- mice. Concomitant nitric oxide synthase inhibition and interleukin-2 supplementation increased the T cell proliferation from both genotypes drastically, but not completely. In sum, we analyzed the involvement of mIDO1 during acute murine toxoplasmosis in our specifically adjusted experimental model and found a definite mIDO1 induction. Nevertheless, mIDO1 seems to be functional redundant as an antiparasitic defense mechanism during acute toxoplasmosis in mice. Furthermore, we suggest that the systemic T cell suppression observed during acute toxoplasmosis is influenced by nitric oxide activity and IL-2 deprivation.


Assuntos
Proliferação de Células/efeitos dos fármacos , Indolamina-Pirrol 2,3,-Dioxigenase/farmacologia , Linfócitos T/efeitos dos fármacos , Toxoplasma/efeitos dos fármacos , Toxoplasmose/tratamento farmacológico , Animais , Modelos Animais de Doenças , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Interleucina-2/metabolismo , Cinurenina/farmacologia , Linfonodos , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico , Óxido Nítrico Sintase/metabolismo , Células RAW 264.7 , RNA Mensageiro/metabolismo , Baço , Toxoplasmose/parasitologia , Transcriptoma , Triptofano/farmacologia
5.
Hepatology ; 70(6): 2075-2091, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31100194

RESUMO

Interleukin-6 (IL-6) is critically involved in liver regeneration after partial hepatectomy (PHX). Previous reports suggest that IL-6 trans-signaling through the soluble IL-6/IL-6R complex is involved in this process. However, the long-term contribution of IL-6 trans-signaling for liver regeneration after PHX is unknown. PHX-induced generation of the soluble IL-6R by ADAM (a disintegrin and metallo) proteases enables IL-6 trans-signaling, in which IL-6 forms an agonistic complex with the soluble IL-6 receptor (sIL-6R) to activate all cells expressing the signal-transducing receptor chain glycoprotein 130 (gp130). In contrast, without activation of ADAM proteases, IL-6 in complex with membrane-bound IL-6R and gp130 activates classic signaling. Here, we describe the generation of IL-6 trans-signaling mice, which exhibit boosted IL-6 trans-signaling and abrogated classic signaling by genetic conversion of all membrane-bound IL-6R into sIL-6R proteins phenocopying hyperactivation of ADAM-mediated shedding of IL-6R as single substrate. Importantly, although IL-6R deficient mice were strongly affected by PHX, survival and regeneration of IL-6 trans-signaling mice was indistinguishable from control mice, demonstrating that IL-6 trans-signaling fully compensates for disabled classic signaling in liver regeneration after PHX. Moreover, we monitored the long-term consequences of global IL-6 signaling inhibition versus IL-6 trans-signaling selective blockade after PHX by IL-6 monoclonal antibodies and soluble glycoprotein 130 as fragment crystallizable fusion, respectively. Both global IL-6 blockade and selective inhibition of IL-6 trans-signaling results in a strong decrease of overall survival after PHX, accompanied by decreased signal transducer and activator of transcription 3 phosphorylation and proliferation of hepatocytes. Mechanistically, IL-6 trans-signaling induces hepatocyte growth factor production by hepatic stellate cells. Conclusion: IL-6 trans-signaling, but not classic signaling, controls liver regeneration following PHX.


Assuntos
Hepatectomia , Interleucina-6/fisiologia , Regeneração Hepática/fisiologia , Animais , Células Estreladas do Fígado/fisiologia , Fator de Crescimento de Hepatócito/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Interleucina-6/sangue , Receptores de Interleucina-6/fisiologia , Transdução de Sinais/fisiologia
6.
Nat Commun ; 10(1): 1739, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30988302

RESUMO

The splenic white pulp is underpinned by poorly characterized stromal cells that demarcate distinct immune cell microenvironments. Here we establish fibroblastic reticular cell (FRC)-specific fate-mapping in mice to define their embryonic origin and differentiation trajectories. Our data show that all reticular cell subsets descend from multipotent progenitors emerging at embryonic day 19.5 from periarterial progenitors. Commitment of FRC progenitors is concluded during the first week of postnatal life through occupation of niches along developing central arterioles. Single cell transcriptomic analysis facilitated deconvolution of FRC differentiation trajectories and indicated that perivascular reticular cells function both as adult lymphoid organizer cells and mural cell progenitors. The lymphotoxin-ß receptor-independent sustenance of postnatal progenitor stemness unveils that systemic immune surveillance in the splenic white pulp is governed through subset specification of reticular cells from a multipotent periarterial progenitor cell. In sum, the finding that discrete signaling events in perivascular niches determine the differentiation trajectories of reticular cell networks explains the development of distinct microenvironmental niches in secondary and tertiary lymphoid tissues that are crucial for the induction and regulation of innate and adaptive immune processes.


Assuntos
Linhagem da Célula , Microambiente Celular , Fibroblastos/fisiologia , Animais , Diferenciação Celular , Perfilação da Expressão Gênica , Vigilância Imunológica , Linfócitos , Camundongos , Baço
7.
Mediators Inflamm ; 2017: 7375818, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28845089

RESUMO

Lymphotoxin ß receptor (LTßR) signaling plays an important role in efficient initiation of host responses to a variety of pathogens, encompassing viruses, bacteria, and protozoans via induction of the type I interferon response. The present study reveals that after Toxoplasma gondii infection, LTßR-/- mice show a substantially reduced survival rate when compared to wild-type mice. LTßR-/- mice exhibit an increased parasite load and a more pronounced organ pathology. Also, a delayed increase of serum IL-12p40 and a failure of the protective IFNγ response in LTßR-/- mice were observed. Serum NO levels in LTßR-/- animals rose later and were markedly decreased compared to wild-type animals. At the transcriptional level, LTßR-/- animals exhibited a deregulated expression profile of several cytokines known to play a role in activation of innate immunity in T. gondii infection. Importantly, expression of the IFNγ-regulated murine guanylate-binding protein (mGBP) genes was virtually absent in the lungs of LTßR-/- mice. This demonstrates clearly that the LTßR is essential for the induction of a type II IFN-mediated immune response against T. gondii. The pronounced inability to effectively upregulate host defense effector molecules such as GBPs explains the high mortality rates of LTßR-/- animals after T. gondii infection.


Assuntos
Interferon gama/metabolismo , Receptor beta de Linfotoxina/metabolismo , Toxoplasma/efeitos dos fármacos , Toxoplasma/metabolismo , Animais , Proteínas de Ligação ao GTP/metabolismo , Imunidade Inata , Subunidade p40 da Interleucina-12/metabolismo , Ketamina/farmacologia , Camundongos , Óxidos de Nitrogênio/sangue , Ligação Proteica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima , Xilazina/farmacologia
8.
J Hepatol ; 64(5): 1108-1117, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26708145

RESUMO

BACKGROUND & AIMS: The liver exhibits a unique capacity for regeneration in response to injury. Lymphotoxin-ß receptor (LTßR), a core member of the tumor necrosis factor (TNF)/tumor necrosis factor receptor (TNFR) superfamily is known to play an important role in this process. However, the function of LTßR during pathophysiological alterations and its molecular mechanisms during liver regeneration are so far ill-characterized. METHODS: LTßR(-/-) mice were subjected to 70% hepatectomy and liver regeneration capacity, bile acid profiles, and transcriptome analysis were performed. RESULTS: LTßR(-/-) deficient mice suffered from increased and prolonged liver tissue damage after 70% hepatectomy, accompanied by deregulated bile acid homeostasis. Pronounced differences in the expression patterns of genes relevant for bile acid synthesis and recirculation were observed. LTßR and TNFRp55 share downstream signalling elements. Therefore, LTßR(-/-) mice were treated with etanercept to create mice functionally deficient in both signalling pathways. Strikingly, the combined blockade of TNFRp55 and LTßR signalling leads to complete failure of liver regeneration resulting in death within 24 to 48h after PHx. Transcriptome analysis revealed a marked disparity in gene expression programs in livers of LTßR(-/-) and etanercept-treated LTßR(-/-) vs. wild-type animals after PHx. Murinoglobulin 2 was identified as a significantly differentially regulated gene. CONCLUSIONS: LTßR is essential for efficient liver regeneration and cooperates with TNFRp55 in this process. Differences in survival kinetics strongly suggest distinct functions for these two cytokine receptors in liver regeneration. Failure of TNFR and LTßR signalling renders liver regeneration impossible.


Assuntos
DNA/genética , Regulação da Expressão Gênica , Hepatopatias/genética , Regeneração Hepática/genética , Receptor beta de Linfotoxina/genética , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Chamariz do Fator de Necrose Tumoral/genética , Animais , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Hepatopatias/metabolismo , Hepatopatias/patologia , Receptor beta de Linfotoxina/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Receptores Tipo I de Fatores de Necrose Tumoral/biossíntese , Receptores Chamariz do Fator de Necrose Tumoral/biossíntese
9.
Nat Immunol ; 13(1): 51-7, 2011 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-22101728

RESUMO

The innate immune system limits viral replication via type I interferon and also induces the presentation of viral antigens to cells of the adaptive immune response. Using infection of mice with vesicular stomatitis virus, we analyzed how the innate immune system inhibits viral propagation but still allows the presentation of antigen to cells of the adaptive immune response. We found that expression of the gene encoding the inhibitory protein Usp18 in metallophilic macrophages led to lower type I interferon responsiveness, thereby allowing locally restricted replication of virus. This was essential for the induction of adaptive antiviral immune responses and, therefore, for preventing the fatal outcome of infection. In conclusion, we found that enforced viral replication in marginal zone macrophages was an immunological mechanism that ensured the production of sufficient antigen for effective activation of the adaptive immune response.


Assuntos
Imunidade Adaptativa , Infecções por Rhabdoviridae/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Replicação Viral/imunologia , Animais , Apresentação de Antígeno/imunologia , Antígenos Virais/imunologia , Linhagem Celular Transformada , Cricetinae , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Endopeptidases/metabolismo , Receptor beta de Linfotoxina/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Imunológicos/metabolismo , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico , Ubiquitina Tiolesterase
10.
Hum Gene Ther ; 22(6): 697-710, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21319998

RESUMO

Gene transfer of mutant O(6)-methylguanine-DNA-methyltransferase (MGMT(P140K)) into hematopoietic stem cells (HSCs) protects hematopoiesis from alkylating agents and allows efficient in vivo selection of transduced HSCs. However, insertional mutagenesis, high regenerative stress associated with selection, and the genotoxic potential of alkylating drugs represent considerable risk factors for clinical applications of this approach. Therefore, we investigated the long-term effect of MGMT(P140K) gene transfer followed by repetitive, dose-intensive treatment with alkylating agents in a murine serial bone marrow transplant model and assessed clonality of hematopoiesis up to tertiary recipients. The substantial selection pressure resulted in almost completely transduced hematopoiesis in all cohorts. Ligation-mediated PCR and next-generation sequencing identified several repopulating clones carrying vector insertions in distinct genomic regions that were ∼ 9 kb of size (common integration sites). Beside polyclonal reconstitution in the majority of the mice, we also detected monoclonal or oligoclonal repopulation patterns with HSC clones showing vector insertions in the Usp10 or Tubb3 gene. Interestingly, neither Usp10, Tubb3, nor any of the genes located in common integration sites have been linked to clonal expansion in previous preclinical or clinical gene therapy trials. However, a considerable number of these genes are involved in DNA damage response and cell fate decision pathways following cytostatic drug application. Thus, in summary, our study advocates ligation-mediated PCR and next generation sequencing as an effective and reliable method to identify gene products associated with clonal survival in specific experimental settings such as chemoselection using alkylating agents.


Assuntos
Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Hematopoese/genética , Células-Tronco Hematopoéticas , Reação em Cadeia da Polimerase/métodos , Análise de Sequência de DNA/métodos , Proteínas Supressoras de Tumor/genética , Alquilantes/farmacologia , Animais , Transplante de Medula Óssea , Células Clonais , Técnicas de Transferência de Genes , Vetores Genéticos , Hematopoese/efeitos dos fármacos , Camundongos , Modelos Animais , Mutação
11.
Mol Ther ; 16(4): 757-64, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18334984

RESUMO

Pulmonary alveolar proteinosis (PAP) due to deficiency of the common beta-chain (beta(c)) of the interleukin-3 (IL-3)/IL-5/granulocyte-macrophage colony-stimulating factor (GM-CSF) receptors is a rare monogeneic disease characterized by functional insufficiency of pulmonary macrophages. Hematopoietic stem cell gene therapy for restoring expression of beta(c)-protein in the hematopoietic system may offer a curative approach. Toward this end, we generated a retroviral construct expressing the murine beta(c) (mbeta(c)) gene and conducted investigations in a murine model of beta(c)-deficient PAP. Functional correction of mbeta(c) activity in mbeta(c)(-/-) bone marrow (BM) cells was demonstrated by restoration of in vitro colony formation in response to GM-CSF. In addition, in a murine in vivo model of mbeta(c)-deficient PAP mbeta(c) gene transfer to hematopoietic stem cells not only restored the GM-CSF-sensitivity of hematopoietic progenitor cells but also, within a period of 12 weeks, almost completely reversed the morphologic features of surfactant accumulation. These results were obtained despite modest transduction levels (10-20%) and, in comparison to wild-type mice, clearly reduced beta(c) expression levels were detected in hematopoietic cells. Therefore, our data demonstrating genetic and functional correction of mbeta(c)(-/-) deficiency in vitro as well as in a murine in vivo model of PAP strongly suggest gene therapy as a potential new treatment modality in beta(c)-deficient PAP.


Assuntos
Subunidade beta Comum dos Receptores de Citocinas/biossíntese , Células-Tronco Hematopoéticas/metabolismo , Proteinose Alveolar Pulmonar/terapia , Animais , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Transplante de Medula Óssea , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Subunidade beta Comum dos Receptores de Citocinas/genética , Terapia Genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Células-Tronco Hematopoéticas/patologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteinose Alveolar Pulmonar/metabolismo , Proteinose Alveolar Pulmonar/patologia , Surfactantes Pulmonares/metabolismo , Retroviridae/genética
12.
Mol Ther ; 16(4): 757-764, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28178466

RESUMO

Pulmonary alveolar proteinosis (PAP) due to deficiency of the common ß-chain (ßc) of the interleukin-3 (IL-3)/IL-5/granulocyte-macrophage colony-stimulating factor (GM-CSF) receptors is a rare monogeneic disease characterized by functional insufficiency of pulmonary macrophages. Hematopoietic stem cell gene therapy for restoring expression of ßc-protein in the hematopoietic system may offer a curative approach. Toward this end, we generated a retroviral construct expressing the murine ßc (mßc) gene and conducted investigations in a murine model of ßc-deficient PAP. Functional correction of mßc activity in mßc-/- bone marrow (BM) cells was demonstrated by restoration of in vitro colony formation in response to GM-CSF. In addition, in a murine in vivo model of mßc-deficient PAP mßc gene transfer to hematopoietic stem cells not only restored the GM-CSF-sensitivity of hematopoietic progenitor cells but also, within a period of 12 weeks, almost completely reversed the morphologic features of surfactant accumulation. These results were obtained despite modest transduction levels (10-20%) and, in comparison to wild-type mice, clearly reduced ßc expression levels were detected in hematopoietic cells. Therefore, our data demonstrating genetic and functional correction of mßc-/- deficiency in vitro as well as in a murine in vivo model of PAP strongly suggest gene therapy as a potential new treatment modality in ßc-deficient PAP.

13.
Cancer Res ; 67(18): 8653-61, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17875705

RESUMO

We here describe a novel xenograft model of chronic lymphocytic leukemia (CLL) generated by infusion of human primary CLL cells into immunodeficient nonobese/severe combined immunodeficient (NOD/SCID) mice. Combined i.v. and i.p. injection of peripheral blood mononuclear cells (PBMC) from 39 patients with CLL resulted in highly reproducible splenic (37 of 39) and peritoneal (35 of 39) engraftment, which remained stable over a time span of 4 to 8 weeks. By comparison, recovery of leukemic cells from bone marrow (21 of 39) or peripheral blood (8 of 22) was substantially lower. The engraftment pattern of CLL PBMC 4 weeks posttransplant was correlated with clinical disease activity: infusion of PBMC from donors with Binet stage A, lymphocyte doubling time of >12 months, and normal lactate dehydrogenase (LDH) serum levels led to marked engraftment of T cells whereas comparably few tumor cells could be detected. In contrast, NOD/SCID mice receiving PBMC from donors with advanced stage Binet C, lymphocyte doubling time of <12 months, and elevated LDH serum levels exhibited predominant engraftment of tumor cells and comparably low numbers of T cells. These results suggest that this model reflects the heterogeneity and important clinical characteristics of the disease, and thus may serve as a tool for preclinical drug testing and investigation of the pathophysiology of CLL.


Assuntos
Modelos Animais de Doenças , Leucemia Linfocítica Crônica de Células B/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Processos de Crescimento Celular/fisiologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Transplante de Neoplasias , Reprodutibilidade dos Testes , Fatores de Risco , Baço/patologia , Linfócitos T/patologia , Transplante Heterólogo
14.
DNA Repair (Amst) ; 6(8): 1197-209, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17499560

RESUMO

As haematopoietic stem cell gene therapy utilizing O(6)-methylguanine-DNA-methyltransferase has reached the clinical stage, safety-related questions become increasingly important. These issues concern insertional mutagenesis of viral vectors, the acute toxicity of pre-transplant conditioning protocols and in vivo selection regimens as well as potential genotoxic side effects of the alkylating drugs administered in this context. To address these questions, we have investigated toxicity-reduced conditioning regimens combining low-dose alkylator application with sublethal irradiation and have analysed their influence on engraftment and subsequent selectability of transduced haematopoietic stem cells. In addition, a strategy to monitor the acute and long-term genotoxic effects of drugs with high guanine-O(6) alkylating potential, such as chloroethylnitrosoureas or temozolomide is introduced. For this purpose, assays were implemented which allow an assessment of the generation and fate of primary drug-induced adducts as well as their long-term effect on chromosomal integrity at the single cell level.


Assuntos
Terapia Genética/efeitos adversos , Terapia Genética/métodos , Células-Tronco Hematopoéticas/enzimologia , O(6)-Metilguanina-DNA Metiltransferase/genética , Substituição de Aminoácidos , Animais , Antineoplásicos Alquilantes/efeitos adversos , Dano ao DNA , Metilases de Modificação do DNA/genética , Reparo do DNA , Enzimas Reparadoras do DNA/genética , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Sítio-Dirigida , Neoplasias/terapia , Proteínas Recombinantes/genética , Segurança , Condicionamento Pré-Transplante , Proteínas Supressoras de Tumor/genética
15.
Intervirology ; 50(3): 197-203, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17283445

RESUMO

OBJECTIVE: Retroviral vectors represent one of the most robust technologies for in vivo expression of heterologous gene sequences and are still the most commonly used vectors in clinical gene therapy trials. The production of high titer retroviral preparations, however, can be a problematic procedure for certain constructs. METHODS: GALV- or RD114-pseudotyped retroviral particles carrying selectable fluorescence markers or drug resistance genes, such as the green fluorescent protein (GFP) or the O(6)-methylguanine-DNA-methyltransferase (MGMT) mutants, were used to stably transduce Phoenix-(FNX-)eco cells. Thereafter, a polyclonal population of producer cells was generated by enriching cells with high marker gene expression. In addition, single producer clones were selected by limiting dilution. RESULTS: Retroviral titers were increased 1-2 logs by enriching for a polyclonal population of producer cells, and selection of single producer clones allowed another 1- to 2-log increase in titers. Using this method, reproducibly high titer viral preparations allowing efficient transduction of hematopoietic stem cells were generated. CONCLUSION: A reliable and time-effective method to generate stable high titer producer cells based on the FNX-cell line for problematic retroviral vector constructs is described.


Assuntos
Linhagem Celular/virologia , Terapia Genética/métodos , Vetores Genéticos , Retroviridae/crescimento & desenvolvimento , Genes Reporter , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Células-Tronco Hematopoéticas , Humanos , O(6)-Metilguanina-DNA Metiltransferase/biossíntese , Reprodutibilidade dos Testes , Transdução Genética
16.
Blood ; 108(9): 2965-71, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16835384

RESUMO

Hematopoietic stem cell gene transfer of the drug-resistance gene cytidine deaminase (CDD) protecting cells from the cytotoxic cytidine analogs cytarabine and gemcitabine was investigated in a murine transplant model. Following transplantation of CDD-transduced cells and cytarabine application (500 mg/kg; days 1-4; intraperitoneally) significant myeloprotection was demonstrated with nadir counts of peripheral blood granulocytes and thrombocytes of 2.9 +/- 0.6/nL versus 0.7 +/- 0.1/nL (P < .001) and 509 +/- 147/nL versus 80 +/- 9/nL (P = .008), respectively (CDD versus control). Protection also was observed from otherwise lethal gemcitabine treatment (250 mg/kg; days 1-3). Stable levels of gene-marked cells in primary and secondary recipients were demonstrated for up to 9 months, and whereas CDD overexpression clearly reduced B- and T-lymphocyte numbers, no major toxicity was observed in the myeloid compartment. Despite the profound myeloprotective properties, however, CDD overexpression did not allow for pharmacologic enrichment of transduced hematopoiesis in our model. Thus, in summary, our data establish CDD as a drug-resistance gene highly suitable for myeloprotective purposes, which, given the lack of selection observed in our hands, might best be used in combination with selectable drug-resistance genes such as MGMT (P140K) or MDR1.


Assuntos
Transplante de Medula Óssea/imunologia , Citarabina/toxicidade , Citidina Desaminase/genética , Desoxicitidina/análogos & derivados , Células 3T3 , Animais , Transplante de Medula Óssea/mortalidade , Ensaio de Unidades Formadoras de Colônias , Citidina Desaminase/metabolismo , Desoxicitidina/toxicidade , Técnicas de Transferência de Genes , Sobrevivência de Enxerto/fisiologia , Contagem de Leucócitos , Camundongos , Modelos Animais , Contagem de Plaquetas , Proteínas Recombinantes de Fusão/metabolismo , Gencitabina
17.
Stem Cells ; 24(3): 722-30, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16195417

RESUMO

For the lymphohematopoietic system, maturation-dependent alterations in DNA repair function have been demonstrated. Because little information is available on the regulatory mechanisms underlying these changes, we have correlated the expression of DNA damage response genes and the functional repair capacity of cells at distinct stages of human hematopoietic differentiation. Comparing fractions of mature (CD34-), progenitor (CD34+ 38+), and stem cells (CD34+ 38-) isolated from umbilical cord blood, we observed: 1) stringently regulated differentiation-dependent shifts in both the cellular processing of DNA lesions and the expression profiles of related genes and 2) considerable interindividual variability of DNA repair at transcriptional and functional levels. The respective repair phenotype was found to be constitutively regulated and not dominated by adaptive response to acute DNA damage. During blood cell development, the removal of DNA adducts, the resealing of repair gaps, the resistance to DNA-reactive drugs clearly increased in stem or mature compared with progenitor cells of the same individual. On the other hand, the vast majority of differentially expressed repair genes was consistently upregulated in the progenitor fraction. A positive correlation of repair function and transcript levels was found for a small number of genes such as RAD23 or ATM, which may serve as key regulators for DNA damage processing via specific pathways. These data indicate that the organism might aim to protect the small number of valuable slow dividing stem cells by extensive DNA repair, whereas fast-proliferating progenitor cells, once damaged, are rather eliminated by apoptosis.


Assuntos
Diferenciação Celular/fisiologia , Reparo do DNA/fisiologia , Sangue Fetal/fisiologia , Regulação da Expressão Gênica/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Linfopoese/fisiologia , ADP-Ribosil Ciclase 1 , Antígenos CD34 , Proliferação de Células , Células Cultivadas , Dano ao DNA , Sangue Fetal/citologia , Perfilação da Expressão Gênica/métodos , Células-Tronco Hematopoéticas/citologia , Humanos , Glicoproteínas de Membrana , Análise de Sequência com Séries de Oligonucleotídeos/métodos
18.
Exp Hematol ; 31(12): 1215-22, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14662327

RESUMO

OBJECTIVE: While retrovirally mediated gene transfer of dihydrofolate reductase mutants (mutDHFR) has convincingly been demonstrated to confer methotrexate (MTX) resistance to murine hematopoietic cells, clinical application of this technology will require high efficacy in human cells. Therefore, we investigated retroviral constructs expressing various point mutants of human DHFR for their ability to confer MTX resistance to human clonogenic progenitor cells (CFU-C) and to allow for in vitro selection of transduced CFU-C. METHODS: Primary human hematopoietic cells were retrovirally transduced using MMLV- and SFFV/MESV-based vectors expressing DHFR(Ser31), DHFR(Phe22/Ser31), or DHFR(Tyr22/Gly31). MTX resistance of unselected and in vitro-selected CFU-C was determined using MTX-supplemented methylcellulose cultures and gene transfer efficiency was assesed by single-colony PCR analysis. RESULTS: While less than 1% mock-transduced CFU-C survived the presence of > or =5 x 10(-8) M MTX, MMLV- and SFFV/MESV-based vectors expressing DHFR(Ser31) significantly protected CFU-C from MTX at doses ranging from 2.5 to 30 x 10(-8) M. Vectors expressing DHFR(Phe22/Ser31) or DHFR(Tyr22/Gly31) were even more protective and MTX-resistant CFU-C were observed up to 1 x 10(-5) M MTX. Three-day suspension cultures in the presence of 10-20 x 10(-8) M MTX resulted in significant selection of mutDHFR-transduced CFU-C. The percentage of CFU-C resistant to 10 x 10(-8) M MTX increased fourfold to 20-fold and provirus-containing CFU-C increased from 27% to 79-100%. CONCLUSION: Gene transfer of DHFR using suitable retroviral backbones and DHFR mutants significantly increases MTX resistance of human CFU-C and allows efficient in vitro selection of transduced cells using a short-term selection procedure.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Células-Tronco Hematopoéticas/metabolismo , Metotrexato/farmacologia , Tetra-Hidrofolato Desidrogenase/genética , Transdução Genética/métodos , Separação Celular/métodos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Vetores Genéticos , Humanos , Mutação Puntual , Tetra-Hidrofolato Desidrogenase/farmacologia , Transdução Genética/normas
19.
Cancer Gene Ther ; 9(9): 737-46, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12189523

RESUMO

The overexpression of mutant forms of O(6)-methylguanine-DNA-methyltransferase (MGMT), resistant to the MGMT inhibitor O(6)-benzylguanine (BG), protects hematopoietic cells from the toxicity of combined BG plus O(6)-alkylating agent chemotherapy. To evaluate the feasibility of this approach for clinically relevant O(6)-alkylating agents, combined therapy with BG and two chloroethylnitrosourea-type drugs, ACNU or BCNU, or the triazene derivative temozolomide (TMZ) was investigated in a murine bone marrow transplant model allowing transgenic expression of the highly BG-resistant MGMT(P140K) mutant. Whereas 20/20 control animals transplanted with nontransduced cells died of progressive myelosuppression during therapy, nearly all animals transplanted with MGMT(P140K)-transduced cells survived treatment with BG/ACNU (12/15), BG/TMZ (10/10), or BG/BCNU (5/5). In surviving animals, hematological parameters improved during chemotherapy and pretreatment levels were reestablished during or shortly after therapy. All animals showed enrichment of transgenic granulocytes (range: 15- to 101-fold) and lymphocytes (range: 16- to 55-fold) in peripheral blood, bone marrow, and spleen. No significant differences were observed between individual treatment groups. Serial transplants demonstrated protection in secondary recipients and confirmed the transduction of transplantable stem cells. Thus, these data demonstrate efficient protection from hematotoxicity and substantial enrichment of transgenic cells following MGMT(P140K) gene transfer and treatment with different O(6)-alkylating drugs.


Assuntos
Células-Tronco Hematopoéticas/enzimologia , O(6)-Metilguanina-DNA Metiltransferase/genética , Transdução Genética , Animais , Antineoplásicos Alquilantes/farmacologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/patologia , Transplante de Medula Óssea , Reparo do DNA , Resistencia a Medicamentos Antineoplásicos , Inibidores Enzimáticos/farmacologia , Feminino , Fluoruracila/farmacologia , Vetores Genéticos/genética , Granulócitos/efeitos dos fármacos , Granulócitos/patologia , Proteínas de Fluorescência Verde , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Interleucina-6/metabolismo , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Retroviridae/genética , Linfócitos T/efeitos dos fármacos , Linfócitos T/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...