Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Gene ; 895: 148018, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-37981082

RESUMO

INTRODUCTION: In contrast to lung adenocarcinoma (LUAD), targetable genetic alterations are less frequently detected in squamous cell carcinoma of the lung (LUSC). Over the last years, gene fusions have become promising targets in many solid cancers. Here, we analysed a cohort of LUSC, identified recurrent fusion genes and functionally characterised these tumour genomes. METHODS: A subset of 1608 squamous cell carcinomas of the lung was analysed by means of the FusionPlex® Lung Panel to identify potentially targetable gene fusions using targeted next-generation sequencing. Cases harbouring recurrent gene fusions were further analysed using FISH, Cytoscan HD arrays and cell culture experiments. RESULTS: We found both, known and novel gene fusions in about 3 % of the cases. Known fusions occurring in lung cancer included ALK::EML4, EGFRvIII, EZR::ROS1 and FGFR3::TACC. We further identified recurrent gene fusions of currently unknown biological function, involving EGFR::VSTM2A and NSD3::FGFR1 and showed that the occurrence of the EGFR::VSTM2A fusion is accompanied by high-level amplification of EGFR. Our analyses further revealed that the genomes of these LUSC patients are chromosomally unstable, which leads us to believe that such non-actionable genomic rearrangements may be a result of "chromosomal chaos" most probably not representing exclusive cancer-driving genes in this cancer entity. CONCLUSIONS: We emphasise that caution should be taken when novel fusions are found and that the appearance of new gene fusions should always be interpreted in the molecular context of the respective disease.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Carcinoma de Células Escamosas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/genética , Quinase do Linfoma Anaplásico/genética , Proteínas Proto-Oncogênicas/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Pulmão/patologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Receptores ErbB/genética , Proteínas de Fusão Oncogênica/genética
2.
Proc Natl Acad Sci U S A ; 120(22): e2211087120, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37216524

RESUMO

Mutations in genes encoding molecular chaperones can lead to chaperonopathies, but none have so far been identified causing congenital disorders of glycosylation. Here we identified two maternal half-brothers with a novel chaperonopathy, causing impaired protein O-glycosylation. The patients have a decreased activity of T-synthase (C1GALT1), an enzyme that exclusively synthesizes the T-antigen, a ubiquitous O-glycan core structure and precursor for all extended O-glycans. The T-synthase function is dependent on its specific molecular chaperone Cosmc, which is encoded by X-chromosomal C1GALT1C1. Both patients carry the hemizygous variant c.59C>A (p.Ala20Asp; A20D-Cosmc) in C1GALT1C1. They exhibit developmental delay, immunodeficiency, short stature, thrombocytopenia, and acute kidney injury (AKI) resembling atypical hemolytic uremic syndrome. Their heterozygous mother and maternal grandmother show an attenuated phenotype with skewed X-inactivation in blood. AKI in the male patients proved fully responsive to treatment with the complement inhibitor Eculizumab. This germline variant occurs within the transmembrane domain of Cosmc, resulting in dramatically reduced expression of the Cosmc protein. Although A20D-Cosmc is functional, its decreased expression, though in a cell or tissue-specific manner, causes a large reduction of T-synthase protein and activity, which accordingly leads to expression of varied amounts of pathological Tn-antigen (GalNAcα1-O-Ser/Thr/Tyr) on multiple glycoproteins. Transient transfection of patient lymphoblastoid cells with wild-type C1GALT1C1 partially rescued the T-synthase and glycosylation defect. Interestingly, all four affected individuals have high levels of galactose-deficient IgA1 in sera. These results demonstrate that the A20D-Cosmc mutation defines a novel O-glycan chaperonopathy and causes the altered O-glycosylation status in these patients.


Assuntos
Injúria Renal Aguda , Chaperonas Moleculares , Masculino , Humanos , Chaperonas Moleculares/metabolismo , Mutação , Polissacarídeos/metabolismo , Células Germinativas/metabolismo
3.
Mol Ther ; 30(2): 593-605, 2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-34678512

RESUMO

Chimeric antigen receptor (CAR)-redirected T cell therapy often fails to control tumors in the long term due to selecting cancer cells that downregulated or lost CAR targeted antigen. To reprogram the functional capacities specifically of engineered CAR T cells, we inserted IL12 into the extracellular moiety of a CD28-ζ CAR; both the CAR endodomain and IL12 were functionally active, as indicated by antigen-redirected effector functions and STAT4 phosphorylation, respectively. The IL12-CAR reprogrammed CD8+ T cells toward a so far not recognized natural killer (NK) cell-like signature and a CD94+CD56+CD62Lhigh phenotype closely similar, but not identical, to NK and cytokine induced killer (CIK) cells. In contrast to conventional CAR T cells, IL12-CAR T cells acquired antigen-independent, human leukocyte antigen E (HLA-E) restricted cytotoxic capacities eliminating antigen-negative cancer cells in addition to eliminating cancer cells with CAR cognate antigen. Simultaneous signaling through both the CAR endodomain and IL12 were required for inducing maximal NK-like cytotoxicity; adding IL12 to conventional CAR T cells was not sufficient. Antigen-negative tumors were attacked by IL12-CAR T cells, but not by conventional CAR T cells. Overall, we present a prototype of a new family of CARs that augments tumor recognition and elimination through expanded functional capacities by an appropriate cytokine integrated into the CAR exodomain.


Assuntos
Linfócitos T CD8-Positivos , Imunoterapia Adotiva , Interleucina-12 , Neoplasias , Linfócitos T CD8-Positivos/imunologia , Humanos , Interleucina-12/imunologia , Células Matadoras Naturais/imunologia , Neoplasias/terapia
4.
Mol Ther ; 27(10): 1825-1835, 2019 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-31331813

RESUMO

Chimeric antigen receptor (CAR)-engineered T cells are efficacious in controlling advanced leukemia and lymphoma, however, they fail in the treatment of solid cancer, which is thought to be due to insufficient T cell activation. We revealed that the immune response of CAR T cells with specificity for carcinoembryonic antigen (CEA) was more efficacious against CEA+ cancer cells when simultaneously incubated with an anti-CD30 immunotoxin or anti-CD30 CAR T cells, although the targeted cancer cells lack CD30. The same effect was achieved when the anti-CD30 single-chain variable fragment (scFv) was integrated into the extracellular domain of the anti-CEA CAR. Improvement in T cell activation was due to interfering with the T cell CD30-CD30L interaction by the antagonistic anti-CD30 scFv HRS3; an agonistic anti-CD30 scFv or targeting the high-affinity interleukin-2 (IL-2) receptor was not effective. T cells with the anti-CD30/CEA CAR showed superior immunity against established CEA+ CD30- tumors in a mouse model. The concept is broadly applicable since anti-CD30/TAG72 CAR T cells also showed improved elimination of TAG72+ CD30- cancer cells. Taken together, targeting CD30 on CAR T cells by the HRS3 scFv within the anti-tumor CAR improves the redirected immune response against solid tumors.


Assuntos
Antígeno Carcinoembrionário/imunologia , Neoplasias do Colo/terapia , Imunoterapia Adotiva/métodos , Antígeno Ki-1/imunologia , Animais , Antígenos de Neoplasias/imunologia , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Células HEK293 , Humanos , Camundongos , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Sci Rep ; 9(1): 8444, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31186450

RESUMO

Little is known on the causes and pathogenesis of the adipose tissue disorder (familial) Multiple Symmetric Lipomatosis (MSL). In a four-generation MSL-family, we performed whole exome sequencing (WES) in 3 affected individuals and 1 obligate carrier and identified Calcyphosine-like (CAPSL) as the most promising candidate gene for this family. Screening of 21 independent patients excluded CAPSL coding sequence variants as a common monogenic cause, but using immunohistochemistry we found that CAPSL was down-regulated in adipose tissue not only from the index patient but also in 10 independent sporadic MSL-patients. This suggests that CAPSL is regulated in sporadic MSL irrespective of the underlying genetic/multifactorial cause. Furthermore, we cultivated pre-adipocytes from MSL-patients and generated 3T3-L1-based Capsl knockout and overexpressing cell models showing altered autophagy, adipogenesis, lipogenesis and Sirtuin-1 (SIRT1) expression. CAPSL seems to be involved in adipocyte biology and perturbation of autophagy is a potential mechanism in the pathogenesis of MSL. Downregulation of CAPSL and upregulation of UCP1 were common features in MSL fat while the known MSL genes MFN2 and LIPE did not show consistent alterations. CAPSL immunostainings could serve as first diagnostic tools in MSL clinical care with a potential to improve time to diagnosis and healthcare options.


Assuntos
Adipogenia/genética , Predisposição Genética para Doença , Lipomatose Simétrica Múltipla/genética , Sirtuína 1/genética , Adipócitos/metabolismo , Adipócitos/patologia , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Idade de Início , Animais , Autofagia/genética , Diferenciação Celular/genética , Feminino , GTP Fosfo-Hidrolases/genética , Regulação da Expressão Gênica/genética , Humanos , Lipomatose Simétrica Múltipla/patologia , Masculino , Camundongos , Proteínas Mitocondriais/genética , Mutação/genética , Linhagem , Sequenciamento do Exoma
7.
Cryobiology ; 84: 10-14, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30148986

RESUMO

Earlier it was shown that number of retrieved follicles was significantly higher in Tumor Dissociation Enzyme (TDE)-treatment group compare to standard Liberase TM-group. The aim of our present investigations was to examine the effect of TDE on appearance of apoptosis and necrosis in follicles and stromal cells after digesting of cryopreserved ovarian cortex. Fresh and frozen ovarian cortex fragments (OCF) from 14 patients (29 ±â€¯6 years old), sized 20-210 mm3 were randomly distributed into four treatment groups and digested with 16% TDE or 0.05 mg/ml Liberase TM: Group 1 frozen OCF digested with TDE; Group 2 frozen OCF digested with LiberaseTM; Group 3 fresh OCF digested with TDE; Group 4 fresh OCF digested with Liberase TM. To differentiate the live, early apoptotic, late apoptotic and necrotic cells in digested ovarian cortex suspension, a flow cytometric apoptosis/necrosis assay with FITC Annexin V Apoptosis Detection Kit and with 7-AAD was performed. Most of fresh (not frozen) cells digested with TDE or Liberase TM (95 ±â€¯2.4% vs. 90.4 ±â€¯3.1%, respectively) as well as in frozen ovarian cortex digested with TDE or Liberase TM (93.1 ±â€¯3.4% vs. 89.7 ±â€¯4.4%, respectively) has located in Q3 quadrant and these cells both negative to 7-AAD and Annexin V were considered as viable. It was established that both types of enzymatic treatment applying to fresh as well as to frozen ovarian cortex resulted to high rate of viable cells (Group 1: 93.8 ±â€¯3.4%; Group 2: 91.8 ±â€¯6.0%; Group 3: 90.5 ±â€¯6.9%; Group 4: 87.3 ±â€¯2.3%) and are non significantly different (P > 0.1) between all treatment groups. The amount of early apoptotic (Group 1: 3.5 ±â€¯1.6%; Group 2: 4.4 ±â€¯1.6%; Group 3: 1.6 ±â€¯1.1%; Group 4: 2.4 ±â€¯1.5%), late apoptotic (Group 1: 2.7 ±â€¯2.4%; Group 2: 44.0 ±â€¯1.9%; Group 3: 3.1 ±â€¯1.1%; Group 4: 2.8 ±â€¯0.7% and necrotic (Group 1: 0.9% ±â€¯0.1%; Group 2: 2.9 ±â€¯0.8%; Group 3: 3.4 ±â€¯4.5%; Group 4: 1.1 ±â€¯0.6%) cells was low and was not significantly different in all treatment groups (P > 0.1). It was concluded that the use of Tumor Dissociation Enzyme, effectiveness of which is higher than Liberase TM, does not lead to increasing of apoptosis and necrosis in follicles and stromal cells after enzymatic digesting of cryopreserved ovarian cortex.


Assuntos
Apoptose , Separação Celular/métodos , Criopreservação/métodos , Preservação da Fertilidade/métodos , Necrose , Folículo Ovariano , Adulto , Animais , Colagenases/farmacologia , Feminino , Humanos , Folículo Ovariano/patologia , Ovário , Termolisina/farmacologia , Adulto Jovem
8.
Reprod Biol Endocrinol ; 16(1): 57, 2018 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-29859539

RESUMO

BACKGROUND: The aim of this study was to examine the effectiveness of Tumor Dissociation Enzyme (TDE) on the viability of follicles after digestion of fresh and cryopreserved ovarian cortex fragments (OCFs). METHODS: Fresh and thawed OCF from 14 patients (29 ± 6 years), sized 20 to 210 mm3 were randomly distributed into four treatment groups and digested with 16% TDE or 0.05 mg/ml Liberase TM: Group 1, frozen OCF digested with TDE; Group 2, frozen OCF digested with LiberaseTM; Group 3, fresh OCF digested with TDE; and Group 4, fresh OCF digested with Liberase TM. Evaluation of follicle viability was performed under light microscope after staining with Neutral red. For visualization of viable and dead cells under a confocal laser scanning microscope, the follicles were stained with Calcein AM and ethidium homodimer-1. RESULTS: The results showed that the number of retrieved follicles was significantly higher (990 vs 487; P < 0.01) in the TDE-treatment group compared to the Liberase TM-group. The presence of intense neutral red stained follicles was significantly higher in Group 1 and Group 3 compared to Group 2 and Group 4 (70.3% ± +/- 6.22 vs 53,1% ± 2.03 and 94.2% ± 6.6 vs 79.1% ± 2.1; P < 0.01). The percentage of Calcein AM stained follicles of class V1 was significantly higher in Group 1 and Group 3 compared to Group 2 and Group 4 (95.97% ± 7.8 vs 87.87% ± 2.4; 97.1% ± 6.8 vs 91.3% ± 2.3; P < 0.01). CONCLUSION: The enzymatic digestion of ovarian cortex with TDE provides recovery of a higher number of healthy preantral follicles in contrast to earlier described Liberase TM procedure.


Assuntos
Colagenases/metabolismo , Criopreservação/métodos , Ovário/enzimologia , Termolisina/metabolismo , Adulto , Sobrevivência Celular/fisiologia , Feminino , Humanos , Microscopia Confocal/métodos , Ovário/citologia , Proteólise , Adulto Jovem
9.
Matrix Biol ; 65: 59-74, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28797711

RESUMO

After skin injury fibroblasts migrate into the wound and transform into contractile, extracellular matrix-producing myofibroblasts to promote skin repair. Persistent activation of myofibroblasts can cause excessive fibrotic reactions, but the underlying mechanisms are not fully understood. We used SMA-GFP transgenic mice to study myofibroblast recruitment and activation in skin wounds. Myofibroblasts were initially recruited to wounds three days post injury, their number reached a maximum after seven days and subsequently declined. Expression profiling showed that 1749 genes were differentially expressed in sorted myofibroblasts from wounds seven days post injury. Most of these genes were linked with the extracellular region and cell periphery including genes encoding for extracellular matrix proteins. A unique panel of core matrisome and matrisome-associated genes was differentially expressed in myofibroblasts and several genes not yet known to be linked to myofibroblast-mediated wound healing were found (e.g. Col24a1, Podnl1, Bvcan, Tinagl1, Thbs3, Adamts16, Adamts19, Cxcl's, Ccl's). In addition, a complex network of G protein-coupled signaling events was regulated in myofibroblasts (e.g. Adcy1, Plbc4, Gnas). Hence, this first characterization of a myofibroblast-specific expression profile at the peak of in situ granulation tissue formation provides important insights into novel target genes that may control excessive ECM deposition during fibrotic reactions.


Assuntos
Actinas/genética , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Pele/lesões , Actinas/metabolismo , Animais , Diferenciação Celular , Modelos Animais de Doenças , Regulação da Expressão Gênica , Camundongos , Camundongos Transgênicos , Miofibroblastos/química , Miofibroblastos/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade de Órgãos , Pele/citologia , Pele/metabolismo
10.
Am J Physiol Lung Cell Mol Physiol ; 313(4): L687-L698, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-28684544

RESUMO

Deficiency of the extracellular matrix protein latent transforming growth factor-ß (TGF-ß)-binding protein-4 (LTBP4) results in lack of intact elastic fibers, which leads to disturbed pulmonary development and lack of normal alveolarization in humans and mice. Formation of alveoli and alveolar septation in pulmonary development requires the concerted interaction of extracellular matrix proteins, growth factors such as TGF-ß, fibroblasts, and myofibroblasts to promote elastogenesis as well as vascular formation in the alveolar septae. To investigate the role of LTBP4 in this context, lungs of LTBP4-deficient (Ltbp4-/-) mice were analyzed in close detail. We elucidate the role of LTBP4 in pulmonary alveolarization and show that three different, interacting mechanisms might contribute to alveolar septation defects in Ltbp4-/- lungs: 1) absence of an intact elastic fiber network, 2) reduced angiogenesis, and 3) upregulation of TGF-ß activity resulting in profibrotic processes in the lung.


Assuntos
Tecido Elástico/patologia , Fibroblastos/patologia , Fibrose/patologia , Proteínas de Ligação a TGF-beta Latente/fisiologia , Pulmão/patologia , Neovascularização Patológica/patologia , Alvéolos Pulmonares/patologia , Animais , Células Cultivadas , Tecido Elástico/metabolismo , Matriz Extracelular/metabolismo , Feminino , Fibroblastos/metabolismo , Fibrose/metabolismo , Pulmão/irrigação sanguínea , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neovascularização Patológica/metabolismo , Organogênese/fisiologia , Alvéolos Pulmonares/metabolismo , Fator de Crescimento Transformador beta/metabolismo
11.
Nat Commun ; 8: 14803, 2017 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-28466852

RESUMO

Low-grade inflammation links obesity to insulin resistance through the activation of tissue-infiltrating immune cells. Interleukin-6 (IL-6) is a crucial regulator of T cells and is increased in obesity. Here we report that classical IL-6 signalling in T cells promotes inflammation and insulin resistance during the first 8 weeks on a high-fat diet (HFD), but becomes dispensable at later stages (after 16 weeks). Mice with T cell-specific deficiency of IL-6 receptor-α (IL-6RαT-KO) exposed to a HFD display improved glucose tolerance, insulin sensitivity and inflammation in liver and EWAT after 8 weeks. However, after 16 weeks, insulin resistance in IL-6RαT-KO epididymal white adipose tissue (EWAT) is comparable to that of controls, whereas the inflammatory profile is significantly worse. This coincided with a shift from classical T cell IL-6 signalling at 8 weeks, to enhanced IL-6 trans-signalling at 16 weeks. Collectively, our studies reveal that IL-6 action in T cells through classical IL-6 signalling promotes inflammation and insulin resistance early during obesity development, which can be compensated for by enhanced IL-6 trans-signalling at later stages.


Assuntos
Inflamação/metabolismo , Resistência à Insulina , Interleucina-6/metabolismo , Obesidade/metabolismo , Transdução de Sinais , Linfócitos T/metabolismo , Animais , Glicemia/metabolismo , Dieta Hiperlipídica , Homeostase , Interleucina-6/genética , Metabolismo dos Lipídeos , Masculino , Camundongos , Camundongos Knockout , Receptores de Interleucina-6/genética , Fatores de Tempo
12.
J Leukoc Biol ; 101(2): 577-587, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27534894

RESUMO

Traditionally, B cells have been best known for their role as producers of antibodies. However, in recent years, a growing body of evidence has accumulated showing that B cells fulfill a range of other immunologic functions. One of the functions that has attracted increasing attention is the capacity of B cells to induce antigen-specific activation of T cells through presentation of antigens. However, the analysis of this B cell function has been hampered by the lack of a phenotypically well-defined antigen-presenting B cell subset. Here, we report the identification of a human antigen-presenting B cell subset with strong immunostimulatory properties. This B cell subset is characterized by low expression of CD21 and high expression of the activation marker CD86 and exhibits strong T cell-stimulatory activity, as demonstrated by means of an autologous mixed-lymphocyte reaction. Phenotypically, CD21lowCD86pos immunostimulatory B cells (BAPC) represented CD27+ class-switched IgMnegIgDneg B lymphocytes and displayed a higher expression of cell surface receptors, which mediate the migration from peripheral blood to sites of inflammation. Flow cytometric analysis of peripheral blood obtained from individuals with inflammatory conditions revealed that the BAPC subset was expanded following vaccination and in patients with rheumatoid arthritis. Taken together, our work shows that BAPC represents a strongly immunostimulatory B cell subset, which could be a promising target for immunotherapeutic intervention in inflammatory diseases.


Assuntos
Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/imunologia , Linfócitos B/imunologia , Linfócitos B/patologia , Inflamação/imunologia , Inflamação/patologia , Adulto , Subpopulações de Linfócitos B/imunologia , Antígeno B7-2/metabolismo , Ligante de CD40/metabolismo , Proliferação de Células , Regulação para Baixo , Humanos , Imunofenotipagem , Ativação Linfocitária/imunologia , Receptores de Antígenos de Linfócitos B/metabolismo , Receptores de Quimiocinas/metabolismo , Receptores de Complemento 3d/metabolismo , Transdução de Sinais , Vacinação
13.
J Cell Biol ; 208(4): 429-42, 2015 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-25688136

RESUMO

Mitochondria form a dynamic network within the cell as a result of balanced fusion and fission. Despite the established role of mitofusins (MFN1 and MFN2) in mitochondrial fusion, only MFN2 has been associated with metabolic and neurodegenerative diseases, which suggests that MFN2 is needed to maintain mitochondrial energy metabolism. The molecular basis for the mitochondrial dysfunction encountered in the absence of MFN2 is not understood. Here we show that loss of MFN2 leads to impaired mitochondrial respiration and reduced ATP production, and that this defective oxidative phosphorylation process unexpectedly originates from a depletion of the mitochondrial coenzyme Q pool. Our study unravels an unexpected and novel role for MFN2 in maintenance of the terpenoid biosynthesis pathway, which is necessary for mitochondrial coenzyme Q biosynthesis. The reduced respiratory chain function in cells lacking MFN2 can be partially rescued by coenzyme Q10 supplementation, which suggests a possible therapeutic strategy for patients with diseases caused by mutations in the Mfn2 gene.


Assuntos
Transporte de Elétrons/genética , GTP Fosfo-Hidrolases/fisiologia , Mitocôndrias/enzimologia , Ubiquinona/análogos & derivados , Trifosfato de Adenosina/biossíntese , Animais , Células Cultivadas , Dinaminas/genética , Metabolismo Energético/genética , Metabolismo Energético/fisiologia , GTP Fosfo-Hidrolases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dinâmica Mitocondrial/fisiologia , Fosforilação Oxidativa , Interferência de RNA , RNA Interferente Pequeno , Terpenos/metabolismo , Ubiquinona/biossíntese
14.
Mol Ther ; 21(12): 2268-77, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23985696

RESUMO

Cytokine-induced killer (CIK) cells raised interest for use in cellular antitumor therapy due to their capability to recognize and destroy autologous tumor cells in a HLA-independent fashion. The antitumor attack of CIK cells, predominantly consisting of terminally differentiated CD8(+)CD56(+) cells, can be improved by redirecting by a chimeric antigen receptor (CAR) that recognizes the tumor cell and triggers CIK cell activation. The requirements for CIK cell activation were, however, so far less explored and are likely to be different from those of "younger" T cells. We revealed that CD28 and OX40 CARs produced higher interferon- secretion as compared with the first-generation ζ-CAR; CD28-ζ and the third-generation CD28-ζ-OX40 CAR, however, performed similar in modulating most CIK cell effector functions. Compared with the CD28-ζ CAR, however, the CD28-ζ-OX40 CAR accelerated terminal maturation of CD56(+) CIK cells producing high frequencies in activation-induced cell death (AICD) and reduced antitumor efficiency in vivo. Consequently, CD28-ζ CAR CIK cells of CD56(-) phenotype were superior in redirected tumor cell elimination. CAR-mediated CIK cell activation also increased antigen-independent target cell lysis; the CD28-ζ CAR was more efficient than the CD28-ζ-OX40 CAR. Translated into therapeutic strategies, CAR-redirected CIK cells benefit from CD28 costimulation; "super-costimulation" by the CD28-ζ-OX40 CAR, however, performed less in antitumor efficacy due to increased AICD.


Assuntos
Antígenos CD28/imunologia , Antígeno CD56/metabolismo , Células Matadoras Induzidas por Citocinas/imunologia , Células Matadoras Induzidas por Citocinas/transplante , Neoplasias Experimentais/terapia , Receptores OX40/imunologia , Animais , Antígenos CD28/metabolismo , Antígeno CD56/imunologia , Morte Celular , Linhagem Celular Tumoral , Células HEK293 , Humanos , Imunoterapia Adotiva , Ativação Linfocitária , Camundongos , Camundongos Knockout , Transplante de Neoplasias , Neoplasias Experimentais/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores OX40/metabolismo , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Linfócitos T/imunologia
15.
Hum Gene Ther ; 24(3): 259-69, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23350854

RESUMO

Adoptive therapy of cancer with genetically redirected T cells showed spectacular efficacy in recent trials. A body of preclinical and clinical data indicate that young effector and central memory T cells perform superior in a primary antitumor response; repetitive antigen engagement, however, drives T-cell maturation to terminally differentiated cells associated with the loss of CCR7, which enables T cells to persist in peripheral tissues. In this work, we explored the antitumor efficacy of CCR7(-) T cells when redirected in an antigen-dependent fashion by a chimeric antigen receptor (CAR) toward tumors in the periphery. CAR-engineered CCR7(-) T cells more efficiently accumulated at the tumor site, secreted more IFN-γ, expressed higher amounts of cytotoxic molecules, and showed superior tumor cell lysis compared to the younger CCR7(+) cells. CCR7(-) T cells, however, were more prone to spontaneous and activation-induced cell death, which could be counteracted by simultaneous CD28 and OX40 (CD134) costimulation. Consequently, the combined CD28-ζ-OX40 signaling CAR rescued CCR7(-) T cells from apoptosis, which then produced more efficient antitumor efficacy than CCR7(+) T cells redirected by the same CAR. Data suggest that T-cell therapy will benefit from combined CD28-ζ-OX40 stimulation in the long-term by rescuing continuously generated CCR7(-) T cells for an antitumor attack.


Assuntos
Antígenos CD28/imunologia , Imunoterapia Adotiva , Receptores CCR7/deficiência , Receptores OX40/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Antígenos CD28/metabolismo , Morte Celular/imunologia , Humanos , Memória Imunológica , Imunofenotipagem , Ativação Linfocitária , Camundongos , Camundongos Knockout , Neoplasias/imunologia , Neoplasias/terapia , Fenótipo , Receptores CCR7/genética , Receptores CCR7/metabolismo , Receptores OX40/genética , Receptores OX40/metabolismo , Transdução de Sinais , Linfócitos T/citologia
16.
PLoS One ; 8(1): e53262, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23308177

RESUMO

Skin injury induces the formation of new blood vessels by activating the vasculature in order to restore tissue homeostasis. Vascular cells may also differentiate into matrix-secreting contractile myofibroblasts to promote wound closure. Here, we characterize a PECAM1(+)/Sca1(+) vascular cell population in mouse skin, which is highly enriched in wounds at the peak of neoangiogenesis and myofibroblast formation. These cells express endothelial and perivascular markers and present the receptor CD38 on their surface. PECAM1(+)/Sca1(+)/CD38(+) cells proliferate upon wounding and could give rise to α-SMA(+) myofibroblast-like cells. CD38 stimulation in immunodeficient mice reduced the wound size at the peak of neoangiogenesis and myofibroblast formation. In humans a corresponding cell population was identified, which was enriched in sprouting vessels of basal cell carcinoma biopsies. The results indicate that PECAM1(+)/Sca1(+)/CD38(+) vascular cells could proliferate and differentiate into myofibroblast-like cells in wound repair. Moreover, CD38 signaling modulates PECAM1(+)/Sca1(+)/CD38(+) cell activation in the healing process implying CD38 as a target for anti-angiogenic therapies in human basal cell carcinoma.


Assuntos
ADP-Ribosil Ciclase 1/metabolismo , Antígenos Ly/metabolismo , Proteínas de Membrana/metabolismo , Miofibroblastos/citologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Fenômenos Fisiológicos da Pele , Pele/irrigação sanguínea , Cicatrização , ADP-Ribosil Ciclase 1/análise , Animais , Antígenos Ly/análise , Carcinoma Basocelular/metabolismo , Prepúcio do Pênis/metabolismo , Prepúcio do Pênis/ultraestrutura , Humanos , Masculino , Proteínas de Membrana/análise , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Miofibroblastos/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise
17.
Gastroenterology ; 143(4): 1095-107.e2, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22750462

RESUMO

BACKGROUND & AIMS: New treatment approaches are needed for patients with pancreatic adenocarcinoma. Carcinoembryonic antigen (CEA) is highly expressed on the surface of pancreatic adenocarcinoma cells; we investigated the effects of cytolytic T cells that recognize CEA in a mouse model of pancreatic carcinoma. METHODS: Immune-competent mice that expressed the CEA transgene (CEAtg) in the intestinal and pulmonary tracts were given intrapancreatic injections of Panc02 CEA(+) cells (express CEA and click beetle luciferase) and tumors were grown for 10 days. Mice were then given single intravenous injections of T cells engineered to express a chimeric antigen receptor (CAR) with high specificity, but moderate affinity, for CEA and a luminescence marker. RESULTS: Injection of the anti-CEA CAR T cells reduced the size of pancreatic tumors to below the limit of detection in all mice and produced long-term tumor eradication in 67% of mice. T cells also eradicated CEA(+) fibrosarcoma cells injected 45 days later. Bioluminescence imaging revealed the accumulation and persistence of the T cells at the tumor site. The efficacy of the T cells did not require lymphodepletion and was not reduced by soluble CEA. Mice developed some noninflammatory infiltrations of CAR(+) T cells in intestine and lung, but there was no evidence of destruction of CEA(+) healthy tissues. CONCLUSIONS: Injection of T cells that target CEA can eradicate tumors grown from CEA(+) pancreatic carcinoma cells in the pancreas of CEAtg mice without autoimmune effects.


Assuntos
Antígeno Carcinoembrionário/imunologia , Antígeno Carcinoembrionário/metabolismo , Carcinoma/terapia , Fibrossarcoma/terapia , Imunoterapia Adotiva , Neoplasias Pancreáticas/terapia , Linfócitos T Citotóxicos/transplante , Animais , Doenças Autoimunes/etiologia , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Carcinoma/metabolismo , Colite/imunologia , Fibrossarcoma/metabolismo , Imunoterapia Adotiva/efeitos adversos , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Pancreáticas/metabolismo , Linfócitos T Citotóxicos/metabolismo
18.
J Bone Miner Res ; 27(11): 2399-412, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22692895

RESUMO

Numerous biochemical studies have pointed to an essential role of annexin A5 (AnxA5), annexin A6 (AnxA6), and collagen X in matrix vesicle-mediated biomineralization during endochondral ossification and in osteoarthritis. By binding to the extracellular matrix protein collagen X and matrix vesicles, annexins were proposed to anchor matrix vesicles in the extracellular space of hypertrophic chondrocytes to initiate the calcification of cartilage. However, mineralization appears to be normal in mice lacking AnxA5 and AnxA6, whereas collagen X-deficient mice show only subtle alterations in the growth plate organization. We hypothesized that the simultaneous lack of AnxA5, AnxA6, and collagen X in vivo induces more pronounced changes in the growth plate development and the initiation of mineralization. In this study, we generated and analyzed mice deficient for AnxA5, AnxA6, and collagen X. Surprisingly, mice were viable, fertile, and showed no obvious abnormalities. Assessment of growth plate development indicated that the hypertrophic zone was expanded in Col10a1(-/-) and AnxA5(-/-) AnxA6(-/-) Col10a1(-/-) newborns, whereas endochondral ossification and mineralization were not affected in 13-day- and 1-month-old mutants. In peripheral quantitative computed tomography, no changes in the degree of biomineralization were found in femora of 1-month- and 1-year-old mutants even though the diaphyseal circumference was reduced in Col10a1(-/-) and AnxA5(-/-) AnxA6(-/-) Col10a1(-/-) mice. The percentage of naive immature IgM(+) /IgM(+) B cells and peripheral T-helper cells were increased in Col10a1(-/-) and AnxA5(-/-) AnxA6(-/-) Col10a1(-/-) mutants, and activated splenic T cells isolated from Col10a1(-/-) mice secreted elevated levels of IL-4 and GM-CSF. Hence, collagen X is needed for hematopoiesis during endochondral ossification and for the immune response, but the interaction of annexin A5, annexin A6, and collagen X is not essential for physiological calcification of growth plate cartilage. Therefore, annexins and collagen X may rather fulfill functions in growth plate cartilage not directly linked to the mineralization process.


Assuntos
Anexina A5/deficiência , Anexina A6/deficiência , Calcificação Fisiológica , Colágeno Tipo X/deficiência , Matriz Extracelular/metabolismo , Hematopoese , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Animais Recém-Nascidos , Anexina A5/metabolismo , Anexina A6/metabolismo , Osso e Ossos/patologia , Colágeno Tipo X/metabolismo , Cruzamentos Genéticos , Feminino , Lâmina de Crescimento/metabolismo , Lâmina de Crescimento/patologia , Hipertrofia , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Mutantes , Células Th1/imunologia , Células Th2/imunologia
19.
PLoS One ; 7(4): e32994, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22539942

RESUMO

The "two-signal paradigm" in T cell activation predicts that the cooperation of "signal 1," provided by the T cell receptor (TCR) through engagement of major histocompatility complex (MHC)-presented peptide, with "signal 2″ provided by costimulatory molecules, the prototype of which is CD28, is required to induce T cell effector functions. While the individual signalling pathways are well understood, little is known about global changes in the proteome pattern during TCR/CD28-mediated activation. Therefore, comparative 2-DE-based proteome analyses of CD3(+) CD69(-) resting T cells versus cells incubated with (i) the agonistic anti-CD3 antibody OKT3 mimicking signal 1 in absence or presence of IL-2 and/or with (ii) the agonistic antibody 15E8 triggering CD28-mediated signaling were performed. Differentially regulated spots were defined leading to the identification of proteins involved in the regulation of the metabolism, shaping and maintenance of the cytoskeleton and signal transduction. Representative members of the differentially expressed protein families, such as calmodulin (CALM), glyceraldehyde-3-phosphate dehydrogenase (GAPDH), L-lactate dehydrogenase (LDH), Rho GDP-dissociation inhibitor 2 (GDIR2), and platelet basic protein (CXCL7), were independently verified by flow cytometry. Data provide a detailed map of individual protein alterations at the global proteome level in response to TCR/CD28-mediated T cell activation.


Assuntos
Proteoma/metabolismo , Linfócitos T/metabolismo , Anticorpos/imunologia , Anticorpos/metabolismo , Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Antígenos CD28/metabolismo , Complexo CD3/metabolismo , Citoesqueleto , Eletroforese em Gel Bidimensional , Citometria de Fluxo , Humanos , Interleucina-2/metabolismo , Lectinas Tipo C/metabolismo , Ativação Linfocitária , Muromonab-CD3/imunologia , Muromonab-CD3/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Linfócitos T/citologia , Linfócitos T/imunologia
20.
PLoS One ; 7(1): e30713, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22292024

RESUMO

Adoptive therapy of malignant diseases with tumor-specific cytotoxic T cells showed remarkable efficacy in recent trials. Repetitive T cell receptor (TCR) engagement of target antigen, however, inevitably ends up in hypo-responsive cells with terminally differentiated KLRG-1(+) CD57(+) CD7(-) phenotype limiting their therapeutic efficacy. We here revealed that hypo-responsiveness of CMV-specific late-stage CD8(+) T cells is due to reduced TCR synapse formation compared to younger cells. Membrane anchoring of TCR components contributes to T cell hypo-responsiveness since dislocation of galectin-3 from the synapse by swainsonine restored both TCR synapse formation and T cell response. Transgenic expression of a CD3-zeta signaling chimeric antigen receptor (CAR) recovered hypo-responsive T cells to full effector functions indicating that the defect is restricted to TCR membrane components while synapse formation of the transgenic CAR was not blocked. CAR engineered late-stage T cells released cytokines and mediated redirected cytotoxicity as efficiently as younger effector T cells. Our data provide a rationale for TCR independent, CAR mediated activation in the adoptive cell therapy to avoid hypo-responsiveness of late-stage T cells upon repetitive antigen encounter.


Assuntos
Complexo CD3/fisiologia , Ativação Linfocitária/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/imunologia , Adulto , Idoso , Complexo CD3/química , Complexo CD3/genética , Complexo CD3/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Células Cultivadas , Infecções por Citomegalovirus/sangue , Infecções por Citomegalovirus/imunologia , Humanos , Ativação Linfocitária/efeitos dos fármacos , Pessoa de Meia-Idade , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/fisiologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Linfócitos T/fisiologia , Transfecção , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...