Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Front Immunol ; 15: 1388272, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38919609

RESUMO

Background: Resection of colorectal liver metastasis is the standard of care for patients with Stage IV CRC. Despite undoubtedly improving the overall survival of patients, pHx for colorectal liver metastasis frequently leads to disease recurrence. The contribution of this procedure to metastatic colorectal cancer at a molecular level is poorly understood. We designed a mouse model of orthograde metastatic colorectal cancer (CRC) to investigate the effect of partial hepatectomy (pHx) on tumor progression. Methods: CRC organoids were implanted into the cecal walls of wild type mice, and animals were screened for liver metastasis. At the time of metastasis, 1/3 partial hepatectomy was performed and the tumor burden was assessed longitudinally using MRI. After euthanasia, different tissues were analyzed for immunological and transcriptional changes using FACS, qPCR, RNA sequencing, and immunohistochemistry. Results: Mice that underwent pHx presented significant liver hypertrophy and an increased overall metastatic load compared with SHAM operated mice in MRI. Elevation in the metastatic volume was defined by an increase in de novo liver metastasis without any effect on the growth of each metastasis. Concordantly, the livers of pHx mice were characterized by neutrophil and bacterial infiltration, inflammatory response, extracellular remodeling, and an increased abundance of tight junctions, resulting in the formation of a premetastatic niche, thus facilitating metastatic seeding. Conclusions: Regenerative pathways following pHx accelerate colorectal metastasis to the liver by priming a premetastatic niche.


Assuntos
Neoplasias Colorretais , Hepatectomia , Neoplasias Hepáticas , Animais , Neoplasias Colorretais/patologia , Camundongos , Neoplasias Hepáticas/secundário , Fígado/patologia , Microambiente Tumoral , Modelos Animais de Doenças , Humanos , Camundongos Endogâmicos C57BL , Inflamação/patologia , Masculino
2.
Angew Chem Int Ed Engl ; 62(36): e202306654, 2023 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-37439488

RESUMO

Metabolic magnetic resonance imaging (MRI) using hyperpolarized (HP) pyruvate is becoming a non-invasive technique for diagnosing, staging, and monitoring response to treatment in cancer and other diseases. The clinically established method for producing HP pyruvate, dissolution dynamic nuclear polarization, however, is rather complex and slow. Signal Amplification By Reversible Exchange (SABRE) is an ultra-fast and low-cost method based on fast chemical exchange. Here, for the first time, we demonstrate not only in vivo utility, but also metabolic MRI with SABRE. We present a novel routine to produce aqueous HP [1-13 C]pyruvate-d3 for injection in 6 minutes. The injected solution was sterile, non-toxic, pH neutral and contained ≈30 mM [1-13 C]pyruvate-d3 polarized to ≈11 % (residual 250 mM methanol and 20 µM catalyst). It was obtained by rapid solvent evaporation and metal filtering, which we detail in this manuscript. This achievement makes HP pyruvate MRI available to a wide biomedical community for fast metabolic imaging of living organisms.


Assuntos
Imageamento por Ressonância Magnética , Ácido Pirúvico , Imageamento por Ressonância Magnética/métodos , Solventes/química , Metanol , Água/química
3.
JCI Insight ; 7(24)2022 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-36413415

RESUMO

Metastatic clear cell renal cell carcinomas (ccRCCs) are resistant to DNA-damaging chemotherapies, limiting therapeutic options for patients whose tumors are resistant to tyrosine kinase inhibitors and/or immune checkpoint therapies. Here we show that mouse and human ccRCCs were frequently characterized by high levels of endogenous DNA damage and that cultured ccRCC cells exhibited intact cellular responses to chemotherapy-induced DNA damage. We identify that pharmacological inhibition of the DNA damage-sensing kinase ataxia telangiectasia and Rad3-related protein (ATR) with the orally administered, potent, and selective drug M4344 (gartisertib) induced antiproliferative effects in ccRCC cells. This effect was due to replication stress and accumulation of DNA damage in S phase. In some cells, DNA damage persisted into subsequent G2/M and G1 phases, leading to the frequent accumulation of micronuclei. Daily single-agent treatment with M4344 inhibited the growth of ccRCC xenograft tumors. M4344 synergized with chemotherapeutic drugs including cisplatin and carboplatin and the poly(ADP-ribose) polymerase inhibitor olaparib in mouse and human ccRCC cells. Weekly M4344 plus cisplatin treatment showed therapeutic synergy in ccRCC xenografts and was efficacious in an autochthonous mouse ccRCC model. These studies identify ATR inhibition as a potential novel therapeutic option for ccRCC.


Assuntos
Antineoplásicos , Carcinoma de Células Renais , Humanos , Animais , Camundongos , Carcinoma de Células Renais/tratamento farmacológico , Cisplatino , Proteínas Mutadas de Ataxia Telangiectasia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico
4.
Nanoscale ; 13(34): 14552-14571, 2021 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-34473175

RESUMO

Iron oxide nanoparticles (IONPs) are well-known contrast agents for MRI for a wide range of sizes and shapes. Their use as theranostic agents requires a better understanding of their magnetic hyperthermia properties and also the design of a biocompatible coating ensuring their stealth and a good biodistribution to allow targeting of specific diseases. Here, biocompatible IONPs of two different shapes (spherical and octopod) were designed and tested in vitro and in vivo to evaluate their abilities as high-end theranostic agents. IONPs featured a dendron coating that was shown to provide anti-fouling properties and a small hydrodynamic size favoring an in vivo circulation of the dendronized IONPs. While dendronized nanospheres of about 22 nm size revealed good combined theranostic properties (r2 = 303 mM s-1, SAR = 395 W gFe-1), octopods with a mean size of 18 nm displayed unprecedented characteristics to simultaneously act as MRI contrast agents and magnetic hyperthermia agents (r2 = 405 mM s-1, SAR = 950 W gFe-1). The extensive structural and magnetic characterization of the two dendronized IONPs reveals clear shape, surface and defect effects explaining their high performance. The octopods seem to induce unusual surface effects evidenced by different characterization techniques while the nanospheres show high internal defects favoring Néel relaxation for magnetic hyperthermia. The study of octopods with different sizes showed that Néel relaxation dominates at sizes below 20 nm while the Brownian one occurs at higher sizes. In vitro experiments demonstrated that the magnetic heating capability of octopods occurs especially at low frequencies. The coupling of a small amount of glucose on dendronized octopods succeeded in internalizing them and showing an effect of MH on tumor growth. All measurements evidenced a particular signature of octopods, which is attributed to higher anisotropy, surface effects and/or magnetic field inhomogeneity induced by tips. This approach aiming at an analysis of the structure-property relationships is important to design efficient theranostic nanoparticles.


Assuntos
Nanopartículas de Magnetita , Medicina de Precisão , Meios de Contraste , Compostos Férricos , Nanopartículas Magnéticas de Óxido de Ferro , Imageamento por Ressonância Magnética , Magnetismo , Nanomedicina Teranóstica , Distribuição Tecidual
5.
Int J Mol Sci ; 22(1)2020 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-33375117

RESUMO

Despite recent advances in therapy, liver metastasis from melanoma is still associated with poor prognosis. Although targeting the mTOR signaling pathway exerts potent anti-tumor activity, little is known about specific mTORC2 inhibition regarding liver metastasis. Using the novel mTORC2 specific inhibitor JR-AB2-011, we show significantly reduced migration and invasion capacity by impaired activation of MMP2 in melanoma cells. In addition, blockade of mTORC2 induces cell death by non-apoptotic pathways and reduces tumor cell proliferation rate dose-dependently. Furthermore, a significant reduction of liver metastasis was detected in a syngeneic murine metastasis model upon therapy with JR-AB2-011 as determined by in vivo imaging and necropsy. Hence, our study for the first time highlights the impact of the pharmacological blockade of mTORC2 as a potent novel anti-cancer approach for liver metastasis from melanoma.


Assuntos
Movimento Celular/efeitos dos fármacos , Neoplasias Hepáticas/prevenção & controle , Alvo Mecanístico do Complexo 2 de Rapamicina/antagonistas & inibidores , Melanoma/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundário , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
6.
Nat Commun ; 11(1): 5133, 2020 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-33046706

RESUMO

Cathepsin D (CTSD) is a lysosomal protease and a marker of poor prognosis in breast cancer. However, the cells responsible for this association and the function of CTSD in cancer are still incompletely understood. By using a conditional CTSD knockout mouse crossed to the transgenic MMTV-PyMT breast cancer model we demonstrate that CTSD deficiency in the mammary epithelium, but not in myeloid cells, blocked tumor development in a cell-autonomous manner. We show that lack of CTSD impaired mechanistic Target of Rapamycin Complex 1 (mTORC1) signaling and induced reversible cellular quiescence. In line, CTSD-deficient tumors started to grow with a two-month delay and quiescent Ctsd-/- tumor cells re-started proliferation upon long-term culture. This was accompanied by rewiring of oncogenic gene expression and signaling pathways, while mTORC1 signaling remained permanently disabled in CTSD-deficient cells. Together, these studies reveal a tumor cell-autonomous effect of CTSD deficiency, and establish a pivotal role of this protease in the cellular response to oncogenic stimuli.


Assuntos
Neoplasias da Mama/metabolismo , Catepsina D/genética , Epitélio/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Animais , Neoplasias da Mama/genética , Catepsina D/deficiência , Feminino , Humanos , Glândulas Mamárias Animais/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais
7.
Recent Results Cancer Res ; 216: 405-437, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32594394

RESUMO

The evolving possibilities of molecular imaging (MI) are fundamentally changing the way we look at cancer, with imaging paradigms now shifting away from basic morphological measures toward the longitudinal assessment of functional, metabolic, cellular, and molecular information in vivo. Recent developments of imaging methodology and probe molecules utilizing the vast number of novel animal models of human cancers have enhanced our ability to non-invasively characterize neoplastic tissue and follow anticancer treatments. While preclinical molecular imaging offers a whole palette of excellent methodology to choose from, we will focus on magnetic resonance imaging (MRI) techniques, since they provide excellent molecular imaging capabilities and bear high potential for clinical translation. Prerequisites and consequences of using animal models as surrogates of human cancers in preclinical molecular imaging are outlined. We present physical principles, values, and limitations of MRI as molecular imaging modality and comment on its high potential to non-invasively assess information on metabolism, hypoxia, angiogenesis, and cell trafficking in preclinical cancer research.


Assuntos
Imageamento por Ressonância Magnética , Oncologia , Imagem Molecular , Neoplasias/diagnóstico por imagem , Animais , Humanos , Neoplasias/metabolismo , Neoplasias/patologia
8.
EMBO J ; 37(15)2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29925518

RESUMO

Polycystic kidney disease (PKD) and other renal ciliopathies are characterized by cysts, inflammation, and fibrosis. Cilia function as signaling centers, but a molecular link to inflammation in the kidney has not been established. Here, we show that cilia in renal epithelia activate chemokine signaling to recruit inflammatory cells. We identify a complex of the ciliary kinase LKB1 and several ciliopathy-related proteins including NPHP1 and PKD1. At homeostasis, this ciliary module suppresses expression of the chemokine CCL2 in tubular epithelial cells. Deletion of LKB1 or PKD1 in mouse renal tubules elevates CCL2 expression in a cell-autonomous manner and results in peritubular accumulation of CCR2+ mononuclear phagocytes, promoting a ciliopathy phenotype. Our findings establish an epithelial organelle, the cilium, as a gatekeeper of tissue immune cell numbers. This represents an unexpected disease mechanism for renal ciliopathies and establishes a new model for how epithelial cells regulate immune cells to affect tissue homeostasis.


Assuntos
Quimiocina CCL2/metabolismo , Cílios/patologia , Doenças Renais Císticas/congênito , Rim Policístico Autossômico Dominante/patologia , Proteína Quinase C/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Quinases Ativadas por AMP , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular , Proteínas do Citoesqueleto , Cães , Células Epiteliais/metabolismo , Feminino , Células HEK293 , Humanos , Doenças Renais Císticas/patologia , Túbulos Renais/citologia , Túbulos Renais/patologia , Macrófagos/metabolismo , Células Madin Darby de Rim Canino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose/fisiologia , Rim Policístico Autossômico Dominante/genética , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Peixe-Zebra
9.
Exp Eye Res ; 166: 168-173, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29074388

RESUMO

The major part of the aqueous humor leaves the eye through the "conventional outflow pathway", consisting of the trabecular meshwork, Schlemm's canal, collector channels, an intrasceral plexus and the episcleral veins. While the trabecular meshwork is well characterized, little is known about anatomical and functional features of the peripheral outflow tract beyond Schlemm's canal. The emergence of minimally-invasive glaucoma surgery directly targeting the outflow resistance in the trabecular meshwork has elicited growing interest in these structures. We used time-of-flight magnetic resonance imaging in ex vivo bovine eyes to map fluid flow under physiological conditions. We were able to identify the peripheral outflow vessels solely by the signal detected from the fluid flow inside their lumina. A question of clinical relevance is, whether localized opening of the trabecular meshwork leads to only localized or to a 360° increase in intrascleral flow. To address this, a goniotomy ab interno was performed in 3 eyes and the flow signal intensity was quantified sectorially. A significant increase in fluid flow was observed in the sector distal to the goniotomy (p = 0.0005) but not in the other sectors (p = 0.1). As a proof of concept we demonstrated that TOF-MRI based detection of flow in the peripheral aqueous outflow tract is feasible. The functional link observed between trabecular meshwork sectors and their distal outflow tract sectors may be relevant for minimally-invasive glaucoma surgery in humans.


Assuntos
Humor Aquoso/metabolismo , Imageamento por Ressonância Magnética/métodos , Tomografia Computadorizada por Raios X/métodos , Animais , Humor Aquoso/diagnóstico por imagem , Bovinos , Modelos Animais de Doenças , Glaucoma/metabolismo , Glaucoma/cirurgia , Malha Trabecular/diagnóstico por imagem , Malha Trabecular/metabolismo , Malha Trabecular/cirurgia
10.
Tumour Biol ; 39(6): 1010428317703922, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28653883

RESUMO

As a potent radiosensitizer nitric oxide (NO) may be a putative adjuvant in the treatment of malignant gliomas which are known for their radio- and chemoresistance. The NO donor prodrug JS-K (O2-(2.4-dinitrophenyl) 1-[(4-ethoxycarbonyl) piperazin-1-yl] diazen-1-ium-1,2-diolate) allows cell-type specific intracellular NO release via enzymatic activation by glutathione-S-transferases overexpressed in glioblastoma multiforme. The cytotoxic and radiosensitizing efficacy of JS-K was assessed in U87 glioma cells in vitro focusing on cell proliferation, induction of DNA damage, and cell death. In vivo efficacy of JS-K and repetitive irradiation were investigated in an orthotopic U87 xenograft model in mice. For the first time, we could show that JS-K acts as a potent cytotoxic and radiosensitizing agent in U87 cells in vitro. This dose- and time-dependent effect is due to an enhanced induction of DNA double-strand breaks leading to mitotic catastrophe as the dominant form of cell death. However, this potent cytotoxic and radiosensitizing effect could not be confirmed in an intracranial U87 xenograft model, possibly due to insufficient delivery into the brain. Although NO donor treatment was well tolerated, neither a retardation of tumor growth nor an extended survival could be observed after JS-K and/or radiotherapy.


Assuntos
Compostos Azo/administração & dosagem , Glioblastoma/tratamento farmacológico , Glioblastoma/radioterapia , Doadores de Óxido Nítrico/administração & dosagem , Piperazinas/administração & dosagem , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Relação Dose-Resposta a Droga , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Glioblastoma/patologia , Humanos , Camundongos , Óxido Nítrico/metabolismo , Radiossensibilizantes/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Nat Cell Biol ; 19(5): 518-529, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28414315

RESUMO

Metastasis is the major cause of cancer-associated death. Partial activation of the epithelial-to-mesenchymal transition program (partial EMT) was considered a major driver of tumour progression from initiation to metastasis. However, the role of EMT in promoting metastasis has recently been challenged, in particular concerning effects of the Snail and Twist EMT transcription factors (EMT-TFs) in pancreatic cancer. In contrast, we show here that in the same pancreatic cancer model, driven by Pdx1-cre-mediated activation of mutant Kras and p53 (KPC model), the EMT-TF Zeb1 is a key factor for the formation of precursor lesions, invasion and notably metastasis. Depletion of Zeb1 suppresses stemness, colonization capacity and in particular phenotypic/metabolic plasticity of tumour cells, probably causing the observed in vivo effects. Accordingly, we conclude that different EMT-TFs have complementary subfunctions in driving pancreatic tumour metastasis. Therapeutic strategies should consider these potential specificities of EMT-TFs to target these factors simultaneously.


Assuntos
Movimento Celular , Plasticidade Celular , Transição Epitelial-Mesenquimal , Neoplasias Pulmonares/metabolismo , Neoplasias Experimentais/metabolismo , Neoplasias Pancreáticas/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo , Animais , Proliferação de Células , Genes p53 , Predisposição Genética para Doença , Proteínas de Homeodomínio/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundário , Camundongos Transgênicos , Mutação , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Fenótipo , Proteínas Proto-Oncogênicas p21(ras)/genética , Interferência de RNA , Transdução de Sinais , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo , Fatores de Tempo , Transativadores/genética , Transfecção , Carga Tumoral , Células Tumorais Cultivadas , Proteína 1 Relacionada a Twist/genética , Proteína 1 Relacionada a Twist/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética
12.
Oncotarget ; 7(51): 85185-85195, 2016 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-27835905

RESUMO

Small cell lung cancer (SCLC) is an aggressive tumor with poor prognosis due to early metastatic spread and development of chemoresistance. Playing a key role in tumor-stroma interactions the CXCL12-CXCR4 axis may be involved in both processes and thus represent a promising therapeutic target in SCLC treatment. In this study we investigated the effect of CXCR4 inhibition on metastasis formation and chemoresistance using an orthotopic xenograft mouse model. This model demonstrates regional spread and spontaneous distant metastases closely reflecting the clinical situation in extensive SCLC. Tumor engraftment, growth, metabolism, and metastatic spread were monitored using different imaging techniques: Magnetic Resonance Imaging (MRI), Bioluminescence Imaging (BLI) and Positron Emission Tomography (PET). Treatment of mice bearing chemoresistant primary tumors with the specific CXCR4 inhibitor AMD3100 reduced the growth of the primary tumor by 61% (P<0.05) and additionally suppressed metastasis formation by 43%. In comparison to CXCR4 inhibition as a monotherapy, standard chemotherapy composed of cisplatin and etoposide reduced the growth of the primary tumor by 71% (P<0.01) but completely failed to suppress metastasis formation. Combination of chemotherapy and the CXCR4 inhibitor integrated the highest of both effects. The growth of the primary tumor was reduced to a similar extent as with chemotherapy alone and metastasis formation was reduced to a similar extent as with CXCR4 inhibitor alone. In conclusion, we demonstrate in this orthotopic mouse model that the addition of a CXCR4 inhibitor to chemotherapy significantly reduces metastasis formation. Thus, it might improve the overall therapy response and consequently the outcome of SCLC patients.


Assuntos
Compostos Heterocíclicos/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Peptídeos/uso terapêutico , Receptores CXCR4/antagonistas & inibidores , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Animais , Benzilaminas , Carcinogênese , Linhagem Celular Tumoral , Movimento Celular , Cisplatino/uso terapêutico , Ciclamos , Proteínas de Ligação a DNA/genética , Resistencia a Medicamentos Antineoplásicos , Etoposídeo/uso terapêutico , Compostos Heterocíclicos/farmacologia , Humanos , Camundongos , Camundongos Knockout , Metástase Neoplásica , Proteínas Nucleares/genética , Peptídeos/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Clin Exp Metastasis ; 33(7): 651-60, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27380917

RESUMO

Small cell lung cancer (SCLC) is a highly aggressive subtype of lung cancer with very poor prognosis due to early metastatic spread and development of chemoresistance. In the last 30 years the study of SCLC has been constrained by a lack of primary human tumor specimen thus highlighting the need of a suitable mouse model. In this article we present the establishment of an orthotopic xenograft mouse model which accurately reproduced the clinical course of SCLC. Orthotopic implantation enabled engraftment of primary lung tumors in all injected mice. Furthermore, immunodeficiency of mice allowed formation of spontaneous metastases in characteristic organs. Bioluminescence Imaging, Magnetic Resonance Imaging and Positron emission tomography were applied to monitor engraftment, metabolism and the exact growth of tumors over time. In order to mimic the extensive disease stage, mice were injected with aggressive human chemoresistant cells leading to development of chemoresistant tumors and early metastatic spread. As a proof of concept treatment of tumor-bearing mice with conventional chemotherapeutics reduced tumor volumes, but a complete regression of tumors was not achieved. By mimicking the extensive disease stage our mouse model can facilitate the study of mechanisms contributing to chemoresistance and metastasis formation, as well as drug screening and evaluation of new treatment strategies for SCLC patients.


Assuntos
Modelos Animais de Doenças , Metástase Neoplásica/patologia , Carcinoma de Pequenas Células do Pulmão/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Pulmão , Imageamento por Ressonância Magnética , Camundongos , Metástase Neoplásica/diagnóstico por imagem , Metástase Neoplásica/genética , Carcinoma de Pequenas Células do Pulmão/diagnóstico por imagem , Carcinoma de Pequenas Células do Pulmão/genética , Transplante Heterólogo/métodos
14.
Theranostics ; 6(6): 862-74, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27162556

RESUMO

Near-infrared photoimmunotherapy (NIR-PIT), which employs monoclonal antibody (mAb)-phototoxic phthalocyanine dye IR700 conjugates, permits the specific, image-guided and spatiotemporally controlled elimination of tumor cells. Here, we report the highly efficient NIR-PIT of human tumor xenografts initiated from patient-derived cancer stem cells (CSCs). Using glioblastoma stem cells (GBM-SCs) expressing the prototypic CSC marker AC133/CD133, we also demonstrate here for the first time that NIR-PIT is highly effective against brain tumors. The intravenously injected theranostic AC133 mAb conjugate enabled the non-invasive detection of orthotopic gliomas by NIR fluorescence imaging, and reached AC133+ GBM-SCs at the invasive tumor front. AC133-targeted NIR-PIT induced the rapid cell death of AC133+ GBM-SCs and thereby strong shrinkage of both subcutaneous and invasively growing brain tumors. A single round of NIR-PIT extended the overall survival of mice with established orthotopic gliomas by more than a factor of two, even though the harmless NIR light was applied through the intact skull. Humanised versions of this theranostic agent may facilitate intraoperative imaging and histopathological evaluation of tumor borders and enable the highly specific and efficient eradication of CSCs.


Assuntos
Antígeno AC133/imunologia , Glioblastoma/diagnóstico por imagem , Glioblastoma/terapia , Imunoterapia/métodos , Fototerapia/métodos , Nanomedicina Teranóstica/métodos , Animais , Anticorpos/administração & dosagem , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/terapia , Modelos Animais de Doenças , Xenoenxertos , Humanos , Indóis/administração & dosagem , Isoindóis , Camundongos , Células-Tronco/imunologia , Análise de Sobrevida , Resultado do Tratamento
15.
MAGMA ; 29(5): 765-75, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27097906

RESUMO

OBJECTIVES: To differentiate between abnormal tumor vessels and regular brain vasculature using new quantitative measures in time-of-flight (TOF) MR angiography (MRA) data. MATERIALS AND METHODS: In this work time-of-flight (TOF) MR angiography data are acquired in 11 glioma patients to quantify vessel abnormality. Brain vessels are first segmented with a new algorithm, efficient monte-carlo image-analysis for the location of vascular entity (EMILOVE), and are then characterized in three brain regions: tumor, normal-appearing contralateral brain, and the total brain volume without the tumor. For characterization local vessel orientation angles and the dot product between local orientation vectors are calculated and averaged in the 3 regions. Additionally, correlation with histological and genetic markers is performed. RESULTS: Both the local vessel orientation angles and the dot product show a statistically significant difference (p < 0.005) between tumor vessels and normal brain vasculature. Furthermore, the connection to both histology and the gene expression of the tumor can be found-here, the measures were compared to the proliferation marker Ki-67 [MIB] and genome-wide expression analysis. The results in a subgroup indicate that the dot product measure may be correlated with activated genetic pathways. CONCLUSION: It is possible to define a measure of vessel abnormality based on local vessel orientation angles which can differentiate between normal brain vasculature and glioblastoma vessels.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/patologia , Glioma/diagnóstico por imagem , Glioma/patologia , Adulto , Idoso , Algoritmos , Encéfalo/diagnóstico por imagem , Estudo de Associação Genômica Ampla , Humanos , Processamento de Imagem Assistida por Computador/métodos , Antígeno Ki-67/metabolismo , Angiografia por Ressonância Magnética , Pessoa de Meia-Idade , Modelos Estatísticos , Método de Monte Carlo , Estudos Retrospectivos
16.
Nat Med ; 20(6): 648-54, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24836575

RESUMO

Acute graft-versus-host disease (GVHD) considerably limits wider usage of allogeneic hematopoietic cell transplantation (allo-HCT). Antigen-presenting cells and T cells are populations customarily associated with GVHD pathogenesis. Of note, neutrophils are the largest human white blood cell population. The cells cleave chemokines and produce reactive oxygen species, thereby promoting T cell activation. Therefore, during an allogeneic immune response, neutrophils could amplify tissue damage caused by conditioning regimens. We analyzed neutrophil infiltration of the mouse ileum after allo-HCT by in vivo myeloperoxidase imaging and found that infiltration levels were dependent on the local microbial flora and were not detectable under germ-free conditions. Physical or genetic depletion of neutrophils reduced GVHD-related mortality. The contribution of neutrophils to GVHD severity required reactive oxygen species (ROS) because selective Cybb (encoding cytochrome b-245, beta polypeptide, also known as NOX2) deficiency in neutrophils impairing ROS production led to lower levels of tissue damage, GVHD-related mortality and effector phenotype T cells. Enhanced survival of Bcl-xL transgenic neutrophils increased GVHD severity. In contrast, when we transferred neutrophils lacking Toll-like receptor-2 (TLR2), TLR3, TLR4, TLR7 and TLR9, which are normally less strongly activated by translocating bacteria, into wild-type C57BL/6 mice, GVHD severity was reduced. In humans, severity of intestinal GVHD strongly correlated with levels of neutrophils present in GVHD lesions. This study describes a new potential role for neutrophils in the pathogenesis of GVHD in both mice and humans.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Íleo/imunologia , Microbiota/imunologia , Neutrófilos/imunologia , Animais , Bussulfano , Ciclofosfamida , Citometria de Fluxo , Adjuvante de Freund , Doença Enxerto-Hospedeiro/fisiopatologia , Técnicas Histológicas , Íleo/microbiologia , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Luciferases , Imageamento por Ressonância Magnética , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise em Microsséries , NADPH Oxidase 2 , NADPH Oxidases/genética , Peroxidase , Espécies Reativas de Oxigênio/metabolismo
17.
J Clin Invest ; 124(7): 2861-76, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24865424

RESUMO

Tissue-specific alternative splicing is critical for the emergence of tissue identity during development, yet the role of this process in malignant transformation is undefined. Tissue-specific splicing involves evolutionarily conserved, alternative exons that represent only a minority of the total alternative exons identified. Many of these conserved exons have functional features that influence signaling pathways to profound biological effect. Here, we determined that lineage-specific splicing of a brain-enriched cassette exon in the membrane-binding tumor suppressor annexin A7 (ANXA7) diminishes endosomal targeting of the EGFR oncoprotein, consequently enhancing EGFR signaling during brain tumor progression. ANXA7 exon splicing was mediated by the ribonucleoprotein PTBP1, which is normally repressed during neuronal development. PTBP1 was highly expressed in glioblastomas due to loss of a brain-enriched microRNA (miR-124) and to PTBP1 amplification. The alternative ANXA7 splicing trait was present in precursor cells, suggesting that glioblastoma cells inherit the trait from a potential tumor-initiating ancestor and that these cells exploit this trait through accumulation of mutations that enhance EGFR signaling. Our data illustrate that lineage-specific splicing of a tissue-regulated alternative exon in a constituent of an oncogenic pathway eliminates tumor suppressor functions and promotes glioblastoma progression. This paradigm may offer a general model as to how tissue-specific regulatory mechanisms can reprogram normal developmental processes into oncogenic ones.


Assuntos
Processamento Alternativo , Anexina A7/genética , Neoplasias Encefálicas/genética , Glioblastoma/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem da Célula/genética , Transformação Celular Neoplásica/genética , Progressão da Doença , Receptores ErbB/genética , Receptores ErbB/metabolismo , Éxons , Técnicas de Silenciamento de Genes , Glioblastoma/metabolismo , Glioblastoma/patologia , Ribonucleoproteínas Nucleares Heterogêneas/antagonistas & inibidores , Ribonucleoproteínas Nucleares Heterogêneas/genética , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neovascularização Patológica/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/antagonistas & inibidores , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Transdução de Sinais/genética , Células Tumorais Cultivadas
18.
J Magn Reson Imaging ; 40(6): 1310-8, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24390982

RESUMO

PURPOSE: Combining multiple imaging biomarkers in one magnetic resonance imaging (MRI) session would be beneficial to gain more data pertaining to tumor vasculature under therapy. Therefore, simultaneous measurement of perfusion, permeability, and vessel size imaging (VSI) using a gradient echo spin echo (GE-SE) sequence with injection of a clinically approved gadolinium (Gd)-based contrast agent was assessed in an orthotopic glioma model. MATERIALS AND METHODS: A combined spin echo gradient echo echo-planar imaging sequence was implemented using a single contrast agent Gd diethylenetriaminepentaacetic acid (Gd-DTPA). This sequence was tested in a mouse brain tumor model (U87_MG), also under treatment with an antiangiogenic agent (bevacizumab). T2 maps and the apparent diffusion coefficient (ADC) were used to differentiate regions of cell death and viable tumor tissue. RESULTS: In viable tumor tissue regional blood volume was 5.7 ± 0.6% in controls and 5.2 ± 0.3% in treated mice. Vessel size was 18.1 ± 2.4 µm in controls and 12.8 ± 2.0 µm in treated mice, which correlated with results from immunohistochemistry. Permeability (K(trans) ) was close to zero in treated viable tumor tissue and 0.062 ± 0.024 min(-1) in controls. CONCLUSION: Our MRI method allows simultaneous assessment of several physiological and morphological parameters and extraction of MRI biomarkers for vasculature. These could be used for treatment monitoring of novel therapeutic agents such as antiangiogenic drugs.


Assuntos
Volume Sanguíneo , Neoplasias Encefálicas/fisiopatologia , Permeabilidade Capilar , Interpretação de Imagem Assistida por Computador/métodos , Imagem Multimodal/métodos , Neovascularização Patológica/fisiopatologia , Animais , Determinação do Volume Sanguíneo/métodos , Neoplasias Encefálicas/complicações , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Imagem Ecoplanar , Feminino , Angiografia por Ressonância Magnética , Camundongos , Camundongos Nus , Neovascularização Patológica/etiologia , Neovascularização Patológica/patologia , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
19.
Nitric Oxide ; 30: 17-25, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23370169

RESUMO

Nitric oxide (NO) released from NO donors can be cytotoxic in tumor cells and can enhance the transport of drugs into brain tumors by altering blood-tumor barrier permeability. The NO donor JS-K [O(2)-(2,4-dinitrophenyl) 1-[(4-ethoxycarbonyl)piperazin-1-yl]diazen-1-ium-1,2-diolate] releases NO upon enzymatic activation selectively in cells overexpressing glutathione-S-transferases (GSTs) such as gliomas. Thus, JS-K-dependent NO effects - especially on cell viability and vascular permeability - were investigated in U87 glioma cells in vitro and in an orthotopic U87 xenograft model in vivo by magnetic resonance imaging (MRI). In vitro experiments showed dose-dependent antiproliferative and cytotoxic effects in U87 cells. In addition, treatment of U87 cells with JS-K resulted in a dose-dependent activation of soluble guanylate cyclase and intracellular accumulation of cyclic guanosine monophosphate (cGMP) which was irreversibly inhibited by the selective inhibitor of soluble guanylate cyclase ODQ (1H-[1,2,4]oxadiazolo(4,3a)quinoxaline-1-one). Using dynamic contrast enhanced MRI (DCE-MRI) as a minimally invasive technique, we demonstrated for the first time a significant increase in the DCE-MRI read-out initial area under the concentration curve (iAUC60) indicating an acute increase in blood-tumor barrier permeability after i.v. treatment with JS-K. Repeated MR imaging of animals with intracranial U87 gliomas under treatment with JS-K (3.5 µmol/kg JS-K 3×/week) and of untreated controls on day 12 and 19 after tumor inoculation revealed no significant changes in tumor growth, edema formation or tumor perfusion. Immunohistochemical workup of the brains showed a significant antiproliferative effect of JS-K in the gliomas. Taken together, in vitro and in vivo data suggest that JS-K has antiproliferative effects in U87 gliomas and opens the blood-tumor barrier by activation of the NO/cGMP signaling pathway. This might be a novel approach to facilitate entry of therapeutic drugs into brain tumors. DCE-MRI is a non-invasive, repeatable imaging modality to monitor biological effects of NO donors and other experimental therapeutics in intracranial tumor models.


Assuntos
Compostos Azo/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Imageamento por Ressonância Magnética/métodos , Doadores de Óxido Nítrico/farmacologia , Piperazinas/farmacologia , Animais , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , GMP Cíclico/metabolismo , Glioma/irrigação sanguínea , Glioma/metabolismo , Humanos , Imuno-Histoquímica , Ratos , Ratos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Immunology ; 137(1): 80-8, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22681312

RESUMO

The therapeutic principle of allogeneic haematopoietic cell transplantation (allo-HCT) is based on an active donor immune system that eliminates host-derived tumour cells. We hypothesized that in addition to the alloantigen-driven anti-tumour response, disruption of the immunological microenvironment within the tumour is responsible for its elimination after allo-HCT. We observed that induction of graft-versus-host disease (GvHD) significantly reduced the abundance of luc(+)  FoxP3(+) regulatory T (Treg) cells in the tumour tissue, which is indicative of impaired or over-ridden tumour recruitment signals towards Treg cells. Analysis of the intestines and liver revealed chemokines and purine nucleotides as candidates for attracting Treg to these sites of inflammation. Despite its expression on tissue-residing Treg cells, the chemokine receptor CCR3 was not critical for Treg-cell function following allo-HCT. Extracellular ATP can attract immune cells via P2Y2. P2Y2 was found to be expressed on Treg cells, and we found a partial reduction of GvHD prevention when P2Y2(-/-) rather than P2Y2(+/+) Treg cells were given. Exogenous local inflammation reduced Treg-cell accumulation in the tumour, suggesting a potential clinical approach to prevent Treg-cell-mediated tumour escape. In conclusion, we demonstrate that GvHD-related inflammation reduced Treg-cell numbers at the tumour sites, which may in turn help to explain the observation that patients with GvHD have a lower risk of tumour relapse.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Linfoma de Células B/imunologia , Receptores Purinérgicos P2Y2/imunologia , Linfócitos T Reguladores/imunologia , Animais , Movimento Celular , Células Cultivadas , Transplante de Células-Tronco Hematopoéticas , Inflamação , Intestinos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Naftalenos/farmacologia , Fenilalanina/análogos & derivados , Fenilalanina/farmacologia , Receptores CCR3/antagonistas & inibidores , Receptores CCR3/metabolismo , Receptores Purinérgicos P2Y2/biossíntese , Receptores Purinérgicos P2Y2/genética , Transdução de Sinais , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/transplante , Transplante Homólogo , Microambiente Tumoral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA