Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Arch Microbiol ; 204(10): 603, 2022 Sep 05.
Article in English | MEDLINE | ID: mdl-36063223

ABSTRACT

Lung cancer, the most prevalent gender-independent tumor entity in both men and women, is among the leading cause of cancer-related deaths worldwide. Despite decades of effort in developing improved therapeutic strategies including immunotherapies and novel chemotherapeutic agents, only modest improvements in outcome and long-term survival of lung cancer patients have been achieved. Therefore, exploring new and exceptional sources for bioactive compounds that might serve as anti-cancer agents might be the key to improving lung cancer therapy. On account of diverse forms, cyanobacteria might serve as a potential source for compounds with potential therapeutic applicability against malignant disorders, including cancer. The assorted arrays of metabolic mechanisms synthesize a plethora of bioactive compounds with immense biological potential. These compounds have been proven to be effective against various cancer cell lines and xenograft animal models. The present review provides an overview of the most promising cyanobacteria-derived bioactive compounds proven to exhibit anti-cancer properties in in-vitro and in-vivo studies and highlights their applicability as potential therapeutic agents with a focus on their anti-lung cancer properties.


Subject(s)
Antineoplastic Agents , Cyanobacteria , Neoplasms , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cyanobacteria/metabolism , Female , Humans
2.
Cell ; 179(7): 1661-1676.e19, 2019 Dec 12.
Article in English | MEDLINE | ID: mdl-31835038

ABSTRACT

Reliable detection of disseminated tumor cells and of the biodistribution of tumor-targeting therapeutic antibodies within the entire body has long been needed to better understand and treat cancer metastasis. Here, we developed an integrated pipeline for automated quantification of cancer metastases and therapeutic antibody targeting, named DeepMACT. First, we enhanced the fluorescent signal of cancer cells more than 100-fold by applying the vDISCO method to image metastasis in transparent mice. Second, we developed deep learning algorithms for automated quantification of metastases with an accuracy matching human expert manual annotation. Deep learning-based quantification in 5 different metastatic cancer models including breast, lung, and pancreatic cancer with distinct organotropisms allowed us to systematically analyze features such as size, shape, spatial distribution, and the degree to which metastases are targeted by a therapeutic monoclonal antibody in entire mice. DeepMACT can thus considerably improve the discovery of effective antibody-based therapeutics at the pre-clinical stage. VIDEO ABSTRACT.


Subject(s)
Antibodies/therapeutic use , Deep Learning , Diagnosis, Computer-Assisted/methods , Drug Therapy, Computer-Assisted/methods , Neoplasms/pathology , Animals , Humans , MCF-7 Cells , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, SCID , Neoplasm Metastasis , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Software , Tumor Microenvironment
3.
Methods Mol Biol ; 1742: 283-300, 2018.
Article in English | MEDLINE | ID: mdl-29330809

ABSTRACT

Solid tumors are often characterized by insufficient oxygen supply (hypoxia), as a result of inadequate vascularization, which cannot keep up with the rapid growth rate of the tumor. Tumor hypoxia is a negative prognostic and predictive factor and is associated with a more aggressive phenotype in various tumor entities. Activation of the hypoxic response in tumors, which is centered around the hypoxia-inducible transcription factors (HIFs), has been causally linked to neovascularization, increased radio- and chemoresistance, altered cell metabolism, genomic instability, increased metastatic potential, and tumor stem cell characteristics. Thus, the hypoxic tumor microenvironment represents a main driving force for tumor progression and a potential target for therapeutic interventions. Here, we describe several methods for the analysis of tumor hypoxia and the hypoxic response in vivo in tumor xenograft models. These methods can be applied to various tumor models, including brain tumor xenotransplants, and allow simultaneously determining the extent and distribution of hypoxia within the tumor, analyzing HIF levels by immunohistochemistry and immunoblot, and quantifying the expression of HIF target genes in tumor tissue. The combination of these approaches provides an important tool to assess the role of the hypoxic tumor microenvironment in vivo.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/surgery , Gene Regulatory Networks , Glioblastoma/surgery , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Hypoxia , Cell Line, Tumor , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Neoplasm Transplantation , Tumor Microenvironment
4.
Cancer Res ; 76(19): 5845-5856, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27488520

ABSTRACT

Hypoxia is a common feature of solid tumors, which controls multiple aspects of cancer progression. One important function of hypoxia and the hypoxia-inducible factors (HIF) is the maintenance of cancer stem-like cells (CSC), a population of tumor cells that possess stem cell-like properties and drives tumor growth. Among the changes promoted by hypoxia is a metabolic shift resulting in acidification of the tumor microenvironment. Here, we show that glioma hypoxia and acidosis functionally cooperate in inducing HIF transcription factors and CSC maintenance. We found that these effects did not involve the classical PHD/VHL pathway for HIF upregulation, but instead involved the stress-induced chaperone protein HSP90. Genetic or pharmacologic inactivation of HSP90 inhibited the increase in HIF levels and abolished the self-renewal and tumorigenic properties of CSCs induced by acidosis. In clinical specimens of glioma, HSP90 was upregulated in the hypoxic niche and was correlated with a CSC phenotype. Our findings highlight the role of tumor acidification within the hypoxic niche in the regulation of HIF and CSC function through HSP90, with implications for therapeutic strategies to target CSC in gliomas and other hypoxic tumors. Cancer Res; 76(19); 5845-56. ©2016 AACR.


Subject(s)
Acidosis/metabolism , Basic Helix-Loop-Helix Transcription Factors/physiology , Brain Neoplasms/pathology , Glioma/pathology , HSP90 Heat-Shock Proteins/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Neoplastic Stem Cells/physiology , Prolyl Hydroxylases/physiology , Von Hippel-Lindau Tumor Suppressor Protein/physiology , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Female , Glioma/metabolism , Humans , Hydrogen-Ion Concentration , Mice
5.
Methods Mol Biol ; 1235: 263-75, 2015.
Article in English | MEDLINE | ID: mdl-25388399

ABSTRACT

Cultured tumor cells are a central tool in cancer research and have provided fundamental insights in tumor biology. Recent evidence, however, indicates that classically established cell lines from different tumors, including glioblastoma, do not fully reflect the genotypes and phenotypes of the respective primary tumors. By contrast, primary cells, isolated from human tumor samples and maintained in serum-free spheroid cultures at low passage under defined growth factor conditions, reproduce key aspects of tumor cell physiology much more faithfully. Among the tumor cell characteristics that are better represented in primary glioblastoma cell cultures is the self-renewal and differentiation potential of the tumor cells. Indeed, a large body of evidence from the past decade indicates that glioblastomas and other tumors are composed of a hierarchy of heterogeneous types of cells, which are generated and maintained by cells that share characteristics of stem cells. This cancer stem cell/tumor initiating cell population is optimally preserved and maintained in primary glioblastoma cultures. Here, we describe a method for the isolation and culture of primary tumor cells from human glioblastomas in serum-free conditions, which allows the routine generation and proper maintenance of tumor cells as spheroid cultures. Such primary tumor cultures can serve as a model of choice for the study of the mechanisms behind key aspects of glioblastoma biology, including tumorigenicity, stem cell hierarchy, invasion, and therapeutic resistance.


Subject(s)
Brain Neoplasms/pathology , Brain/cytology , Brain/pathology , Cell Culture Techniques/methods , Cell Separation/methods , Glioblastoma/pathology , Cryopreservation/methods , Humans , Neoplastic Stem Cells/pathology , Spheroids, Cellular , Tumor Cells, Cultured
6.
Nat Commun ; 5: 5577, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25420589

ABSTRACT

Tumours exploit their hypoxic microenvironment to induce a more aggressive phenotype, while curtailing the growth-inhibitory effects of hypoxia through mechanisms that are poorly understood. The prolyl hydroxylase PHD3 is regulated by hypoxia and plays an important role in tumour progression. Here we identify PHD3 as a central regulator of epidermal growth factor receptor (EGFR) activity through the control of EGFR internalization to restrain tumour growth. PHD3 controls EGFR activity by acting as a scaffolding protein that associates with the endocytic adaptor Eps15 and promotes the internalization of EGFR. In consequence, loss of PHD3 in tumour cells suppresses EGFR internalization and hyperactivates EGFR signalling to enhance cell proliferation and survival. Our findings reveal that PHD3 inactivation provides a novel route of EGFR activation to sustain proliferative signalling in the hypoxic microenvironment.


Subject(s)
Endocytosis , ErbB Receptors/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Neoplasms/enzymology , Signal Transduction , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Cell Line, Tumor , Cell Proliferation , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Neoplasms/genetics , Neoplasms/physiopathology , Protein Binding
7.
Nat Commun ; 5: 5582, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25420773

ABSTRACT

Solid tumours are exposed to microenvironmental factors such as hypoxia that normally inhibit cell growth. However, tumour cells are capable of counteracting these signals through mechanisms that are largely unknown. Here we show that the prolyl hydroxylase PHD3 restrains tumour growth in response to microenvironmental cues through the control of EGFR. PHD3 silencing in human gliomas or genetic deletion in a murine high-grade astrocytoma model markedly promotes tumour growth and the ability of tumours to continue growing under unfavourable conditions. The growth-suppressive function of PHD3 is independent of the established PHD3 targets HIF and NF-κB and its hydroxylase activity. Instead, loss of PHD3 results in hyperphosphorylation of epidermal growth factor receptor (EGFR). Importantly, epigenetic/genetic silencing of PHD3 preferentially occurs in gliomas without EGFR amplification. Our findings reveal that PHD3 inactivation provides an alternative route of EGFR activation through which tumour cells sustain proliferative signalling even under conditions of limited oxygen availability.


Subject(s)
Cell Proliferation , ErbB Receptors/metabolism , Glioblastoma/physiopathology , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia/physiopathology , Procollagen-Proline Dioxygenase/genetics , Animals , Apoptosis , Cell Line, Tumor , ErbB Receptors/genetics , Female , Gene Knockout Techniques , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Hypoxia/enzymology , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Male , Mice, Knockout , Oxygen/metabolism , Procollagen-Proline Dioxygenase/deficiency
8.
J Exp Med ; 209(9): 1611-27, 2012 Aug 27.
Article in English | MEDLINE | ID: mdl-22908324

ABSTRACT

Endothelial Wnt/ß-catenin signaling is necessary for angiogenesis of the central nervous system and blood-brain barrier (BBB) differentiation, but its relevance for glioma vascularization is unknown. In this study, we show that doxycycline-dependent Wnt1 expression in subcutaneous and intracranial mouse glioma models induced endothelial Wnt/ß-catenin signaling and led to diminished tumor growth, reduced vascular density, and normalized vessels with increased mural cell attachment. These findings were corroborated in GL261 glioma cells intracranially transplanted in mice expressing dominant-active ß-catenin specifically in the endothelium. Enforced endothelial ß-catenin signaling restored BBB characteristics, whereas inhibition by Dkk1 (Dickkopf-1) had opposing effects. By overactivating the Wnt pathway, we induced the Wnt/ß-catenin-Dll4/Notch signaling cascade in tumor endothelia, blocking an angiogenic and favoring a quiescent vascular phenotype, indicated by induction of stalk cell genes. We show that ß-catenin transcriptional activity directly regulated endothelial expression of platelet-derived growth factor B (PDGF-B), leading to mural cell recruitment thereby contributing to vascular quiescence and barrier function. We propose that reinforced Wnt/ß-catenin signaling leads to inhibition of angiogenesis with normalized and less permeable vessels, which might prove to be a valuable therapeutic target for antiangiogenic and edema glioma therapy.


Subject(s)
Central Nervous System Neoplasms/blood supply , Central Nervous System Neoplasms/metabolism , Glioma/blood supply , Glioma/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing , Animals , Blood-Brain Barrier/metabolism , Brain Neoplasms/blood supply , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Calcium-Binding Proteins , Central Nervous System Neoplasms/pathology , Endothelium, Vascular/metabolism , Female , Forkhead Transcription Factors/genetics , Glioma/pathology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Nude , Neoplasm Grading , Neovascularization, Pathologic , Proto-Oncogene Proteins c-sis/genetics , Wnt1 Protein/genetics , Wnt1 Protein/metabolism , Xenograft Model Antitumor Assays
9.
Nature ; 465(7297): 487-91, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20445540

ABSTRACT

The formation and guidance of specialized endothelial tip cells is essential for both developmental and pathological angiogenesis. Notch-1 signalling regulates the generation of tip cells, which respond to gradients of vascular endothelial growth factor (VEGF-A). The molecular cues and signalling pathways that control the guidance of tip cells are poorly understood. Bidirectional signalling by Eph receptors and ephrin ligands represents one of the most important guidance cues involved in axon path finding. Here we show that ephrin-B2 reverse signalling involving PDZ interactions regulates endothelial tip cell guidance to control angiogenic sprouting and branching in physiological and pathological angiogenesis. In vivo, ephrin-B2 PDZ-signalling-deficient mice (ephrin-B2DeltaV) exhibit a reduced number of tip cells with fewer filopodial extensions at the vascular front in the mouse retina. In pathological settings, impaired PDZ signalling decreases tumour vascularization and growth. Mechanistically, we show that ephrin-B2 controls VEGF receptor (VEGFR)-2 internalization and signalling. Importantly, internalization of VEGFR2 is necessary for activation and downstream signalling of the receptor and is required for VEGF-induced tip cell filopodial extension. Together, our results suggest that ephrin-B2 at the tip cell filopodia regulates the proper spatial activation of VEGFR2 endocytosis and signalling to direct filopodial extension. Blocking ephrin-B2 reverse signalling may be an attractive alternative or combinatorial anti-angiogenic therapy strategy to disrupt VEGFR2 function in tumour angiogenesis.


Subject(s)
Astrocytoma/blood supply , Astrocytoma/metabolism , Ephrin-B2/metabolism , Neovascularization, Pathologic , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Astrocytoma/pathology , Brain/blood supply , Cells, Cultured , Endocytosis , Endothelial Cells/cytology , Endothelial Cells/metabolism , Ephrin-B2/deficiency , Ephrin-B2/genetics , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neovascularization, Physiologic , Pseudopodia/metabolism , Retina , Retinal Vessels/cytology , Retinal Vessels/physiology , Signal Transduction
10.
Brain ; 133(Pt 4): 983-95, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20375133

ABSTRACT

Glioma growth and progression depend on a specialized subpopulation of tumour cells, termed tumour stem cells. Thus, tumour stem cells represent a critical therapeutic target, but the molecular mechanisms that regulate them are poorly understood. Hypoxia plays a key role in tumour progression and in this study we provide evidence that the hypoxic tumour microenvironment also controls tumour stem cells. We define a detailed molecular signature of tumour stem cell genes, which are overexpressed by tumour cells in vascular and perinecrotic/hypoxic niches. Mechanistically, we show that hypoxia plays a key role in the regulation of the tumour stem cell phenotype through hypoxia-inducible factor 2alpha and subsequent induction of specific tumour stem cell signature genes, including mastermind-like protein 3 (Notch pathway), nuclear factor of activated T cells 2 (calcineurin pathway) and aspartate beta-hydroxylase domain-containing protein 2. Notably, a number of these genes belong to pathways regulating the stem cell phenotype. Consistently, tumour stem cell signature genes are overexpressed in newly formed gliomas and are associated with worse clinical prognosis. We propose that tumour stem cells are maintained within a hypoxic niche, providing a functional link between the well-established role of hypoxia in stem cell and tumour biology. The identification of molecular regulators of tumour stem cells in the hypoxic niche points to specific signalling mechanisms that may be used to target the glioblastoma stem cell population.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Glioblastoma/metabolism , Glioblastoma/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Cell Hypoxia/physiology , Cell Line, Tumor , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques/methods , Humans
SELECTION OF CITATIONS
SEARCH DETAIL