Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters

Publication year range
1.
Physiol Rev ; 101(2): 569-610, 2021 04 01.
Article in English | MEDLINE | ID: mdl-32730114

ABSTRACT

Fibroblast growth factors (FGFs) are a family of proteins possessing paracrine, autocrine, or endocrine functions in a variety of biological processes, including embryonic development, angiogenesis, tissue homeostasis, wound repair, and cancer. Canonical FGFs bind and activate tyrosine kinase FGF receptors (FGFRs), triggering intracellular signaling cascades that mediate their biological activity. Experimental evidence indicates that FGFs play a complex role in the physiopathology of the prostate gland that ranges from essential functions during embryonic development to modulation of neoplastic transformation. The use of ligand- and receptor-deleted mouse models has highlighted the requirement for FGF signaling in the normal development of the prostate gland. In adult prostate, the maintenance of a functional FGF/FGFR signaling axis is critical for organ homeostasis and function, as its disruption leads to prostate hyperplasia and may contribute to cancer progression and metastatic dissemination. Dissection of the molecular landscape modulated by the FGF family will facilitate ongoing translational efforts directed toward prostate cancer therapy.


Subject(s)
Fibroblast Growth Factors/physiology , Prostate/physiology , Prostate/physiopathology , Prostatic Diseases/physiopathology , Prostatic Neoplasms/physiopathology , Receptors, Fibroblast Growth Factor/physiology , Animals , Humans , Intercellular Signaling Peptides and Proteins/physiology , Male , Prostate/growth & development
2.
Pharmacol Res ; 206: 107291, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38969274

ABSTRACT

Fibroblast growth factors (FGFs) act as proangiogenic and mitogenic cytokines in several cancers, including multiple myeloma (MM). Indeed, corrupted FGF autocrine and paracrine secretion induces an aberrant activation of the FGF receptor (FGFR) signaling sustaining cancer cell spreading and resistance to pharmacological treatments. Thus, FGF traps may represent a promising anti-cancer strategy to hamper the ligand-dependent activation of the FGF/FGFR system. We previously identified NSC12 as the first orally available small molecule FGF trap able to inhibit the growth and progression of several FGF-dependent tumor models. NSC12 is a pregnenolone derivative carrying a 1,1-bis-trifluoromethyl-1,3-propanediol chain in position 17 of the steroid nucleus. Investigation of structure-activity relationships (SARs) provided more potent and specific NSC12 steroid derivatives and highlighted that the C17-side chain is pivotal for the FGF trap activity. Here, a scaffold hopping approach allowed to obtain two FGF trap compounds (22 and 57) devoid of the steroid nucleus and able to efficiently bind FGF2 and to inhibit FGFR activation in MM cells. Accordingly, these compounds exert a potent anti-tumor activity on MM cell lines both in vitro and in vivo and on MM patient-derived primary cells, strongly affecting the survival of both proteasome-inhibitor sensitive and resistant MM cells. These results propose a new therapeutic option for relapsed/refractory MM patients and set the bases for the development of novel FGF traps prone to chemical diversification to be used in the clinic for the treatment of those tumors in which the FGF/FGFR system plays a pivotal role, including MM.


Subject(s)
Antineoplastic Agents , Fibroblast Growth Factors , Multiple Myeloma , Receptors, Fibroblast Growth Factor , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Humans , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Cell Line, Tumor , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/metabolism , Fibroblast Growth Factors/metabolism , Structure-Activity Relationship , Drug Discovery , Mice , Fibroblast Growth Factor 2/metabolism
3.
Blood ; 137(18): 2495-2508, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33197938

ABSTRACT

The human fibroblast growth factor/fibroblast growth factor receptor (FGF/FGFR) axis deregulation is largely involved in supporting the pathogenesis of hematologic malignancies, including Waldenström macroglobulinemia (WM). WM is still an incurable disease, and patients succumb because of disease progression. Therefore, novel therapeutics designed to specifically target deregulated signaling pathways in WM are required. We aimed to investigate the role of FGF/FGFR system blockade in WM by using a pan-FGF trap molecule (NSC12). Wide-transcriptome profiling confirmed inhibition of FGFR signaling in NSC12-treated WM cells; unveiling a significant inhibition of MYD88 was also confirmed at the protein level. Importantly, the NSC12-dependent silencing of MYD88 was functionally active, as it led to inhibition of MYD88-driven pathways, such as BTK and SYK, as well as the MYD88-downstream target HCK. Of note, both canonical and noncanonical NF-κB cascades were downregulated in WM cells upon NSC12 treatment. Functional sequelae exerted by NSC12 in WM cells were studied, demonstrating significant inhibition of WM cell growth, induction of WM cell apoptosis, halting MAPK, JAK/STAT3, and PI3K-Akt pathways. Importantly, NSC12 exerted an anti-WM effect even in the presence of bone marrow microenvironment, both in vitro and in vivo. Our studies provide the evidence for using NSC12 as a specific FGF/FGFR system inhibitor, thus representing a novel therapeutic strategy in WM.


Subject(s)
Biomarkers, Tumor/metabolism , Cholesterol/analogs & derivatives , Fibroblast Growth Factors/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Myeloid Differentiation Factor 88/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Waldenstrom Macroglobulinemia/prevention & control , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Cholesterol/pharmacology , Gene Expression Profiling , Humans , Mice , Signal Transduction , Tumor Cells, Cultured , Tumor Microenvironment , Waldenstrom Macroglobulinemia/genetics , Waldenstrom Macroglobulinemia/metabolism , Waldenstrom Macroglobulinemia/pathology , Xenograft Model Antitumor Assays
4.
Blood ; 138(18): 1705-1720, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34077955

ABSTRACT

Alterations in KRAS have been identified as the most recurring somatic variants in the multiple myeloma (MM) mutational landscape. Combining DNA and RNA sequencing, we studied 756 patients and observed KRAS as the most frequently mutated gene in patients at diagnosis; in addition, we demonstrated the persistence or de novo occurrence of the KRAS aberration at disease relapse. Small-molecule inhibitors targeting KRAS have been developed; however, they are selective for tumors carrying the KRASG12C mutation. Therefore, there is still a need to develop novel therapeutic approaches to target the KRAS mutational events found in other tumor types, including MM. We used AZD4785, a potent and selective antisense oligonucleotide that selectively targets and downregulates all KRAS isoforms, as a tool to dissect the functional sequelae secondary to KRAS silencing in MM within the context of the bone marrow niche and demonstrated its ability to significantly silence KRAS, leading to inhibition of MM tumor growth, both in vitro and in vivo, and confirming KRAS as a driver and therapeutic target in MM.


Subject(s)
Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Mutation/drug effects , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Humans , Mice, SCID , Molecular Targeted Therapy , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Oligonucleotides, Antisense/therapeutic use , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use
5.
Cancer Cell Int ; 23(1): 89, 2023 May 11.
Article in English | MEDLINE | ID: mdl-37165394

ABSTRACT

BACKGROUND: Cancer stem-like cells (CSCs) are a subpopulation of tumor cells responsible for tumor initiation, metastasis, chemoresistance, and relapse. Recently, CSCs have been identified in Uveal Melanoma (UM), which represents the most common primary tumor of the eye. UM is highly resistant to systemic chemotherapy and effective therapies aimed at improving overall survival of patients are eagerly required. METHODS: Herein, taking advantage from a pan Fibroblast Growth Factor (FGF)-trap molecule, we singled out and analyzed a UM-CSC subset with marked stem-like properties. A hierarchical clustering of gene expression data publicly available on The Cancer Genome Atlas (TCGA) was performed to identify patients' clusters. RESULTS: By disrupting the FGF/FGF receptor (FGFR)-mediated signaling, we unmasked an FGF-sensitive UM population characterized by increased expression of numerous stemness-related transcription factors, enhanced aldehyde dehydrogenase (ALDH) activity, and tumor-sphere formation capacity. Moreover, FGF inhibition deeply affected UM-CSC survival in vivo in a chorioallantoic membrane (CAM) tumor graft assay, resulting in the reduction of tumor growth. At clinical level, hierarchical clustering of TCGA gene expression data revealed a strong correlation between FGFs/FGFRs and stemness-related genes, allowing the identification of three distinct clusters characterized by different clinical outcomes. CONCLUSIONS: Our findings support the evidence that the FGF/FGFR axis represents a master regulator of cancer stemness in primary UM tumors and point to anti-FGF treatments as a novel therapeutic strategy to hit the CSC component in UM.

6.
Pharmacol Res ; 189: 106683, 2023 03.
Article in English | MEDLINE | ID: mdl-36736415

ABSTRACT

In spite of the huge advancements in both diagnosis and interventions, hormone refractory prostate cancer (HRPC) remains a major hurdle in prostate cancer (PCa). Metabolic reprogramming plays a key role in PCa oncogenesis and resistance. However, the dynamics between metabolism and oncogenesis are not fully understood. Here, we demonstrate that two multi-target natural products, cannabidiol (CBD) and cannabigerol (CBG), suppress HRPC development in the TRansgenic Adenocarcinoma of the Mouse Prostate (TRAMP) model by reprogramming metabolic and oncogenic signaling. Mechanistically, CBD increases glycolytic capacity and inhibits oxidative phosphorylation in enzalutamide-resistant HRPC cells. This action of CBD originates from its effect on metabolic plasticity via modulation of VDAC1 and hexokinase II (HKII) coupling on the outer mitochondrial membrane, which leads to strong shifts of mitochondrial functions and oncogenic signaling pathways. The effect of CBG on enzalutamide-resistant HRPC cells was less pronounced than CBD and only partially attributable to its action on mitochondria. However, when optimally combined, these two cannabinoids exhibited strong anti-tumor effects in TRAMP mice, even when these had become refractory to enzalutamide, thus pointing to their therapeutical potential against PCa.


Subject(s)
Cannabidiol , Prostatic Neoplasms , Humans , Male , Mice , Animals , Cannabidiol/pharmacology , Cell Death , Mitochondria/metabolism , Prostatic Neoplasms/metabolism , Oxidative Phosphorylation , Carcinogenesis/metabolism , Hormones/metabolism , Voltage-Dependent Anion Channel 1/metabolism
7.
Biochim Biophys Acta Rev Cancer ; 1869(1): 53-63, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29175552

ABSTRACT

Since its discovery in 1992, long pentraxin 3 (PTX3) has been characterized as soluble patter recognition receptor, a key player of the innate immunity arm with non-redundant functions in pathogen recognition and inflammatory responses. As a component of the extra-cellular matrix milieu, PTX3 has been implicated also in wound healing/tissue remodeling, cardiovascular diseases, fertility, and infectious diseases. Consequently, PTX3 levels in biological fluids have been proposed as a fluid-phase biomarker in different pathological conditions. In the last decade, experimental evidences have shown that PTX3 may exert a significant impact also on different aspects of cancer biology, including tumor onset, angiogenesis, metastatic dissemination and immune-modulation. However, it remains unclear whether PTX3 acts as a good cop or bad cop in cancer. In this review, we will summarize and discuss the scientific literature data focusing on the role of PTX3 in experimental and human tumors, including its putative translational implications.


Subject(s)
C-Reactive Protein/physiology , Neoplasms/genetics , Serum Amyloid P-Component/physiology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , C-Reactive Protein/genetics , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Gene Expression Regulation, Neoplastic , Humans , Inflammation/genetics , Inflammation/metabolism , Neoplasms/pathology , Protein Isoforms/genetics , Protein Isoforms/physiology , Serum Amyloid P-Component/genetics
8.
Int J Mol Sci ; 21(24)2020 Dec 09.
Article in English | MEDLINE | ID: mdl-33317057

ABSTRACT

Lung cancer represents an extremely diffused neoplastic disorder with different histological/molecular features. Among the different lung tumors, non-small-cell lung cancer (NSCLC) is the most represented histotype, characterized by various molecular markers, including the expression/overexpression of the fibroblast growth factor receptor-1 (FGFR1). Thus, FGF/FGFR blockade by tyrosine kinase inhibitors (TKi) or FGF-ligand inhibitors may represent a promising therapeutic approach in lung cancers. In this study we demonstrate the potential therapeutic benefit of targeting the FGF/FGFR system in FGF-dependent lung tumor cells using FGF trapping (NSC12) or TKi (erdafitinib) approaches. The results show that inhibition of FGF/FGFR by NSC12 or erdafitinib induces apoptosis in FGF-dependent human squamous cell carcinoma NCI-H1581 and NCI-H520 cells. Induction of oxidative stress is the main mechanism responsible for the therapeutic/pro-apoptotic effect exerted by both NSC12 and erdafitinib, with apoptosis being abolished by antioxidant treatments. Finally, reduction of c-Myc protein levels appears to strictly determine the onset of oxidative stress and the therapeutic response to FGF/FGFR inhibition, indicating c-Myc as a key downstream effector of FGF/FGFR signaling in FGF-dependent lung cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Lung Neoplasms/metabolism , Oxidative Stress , Protein Kinase Inhibitors/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cholesterol/analogs & derivatives , Cholesterol/pharmacology , Cholesterol/therapeutic use , Down-Regulation , Female , Fibroblast Growth Factors/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mice , Mice, Inbred C57BL , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Quinoxalines/pharmacology , Quinoxalines/therapeutic use , Receptors, Fibroblast Growth Factor/metabolism
9.
Int J Mol Sci ; 20(10)2019 May 14.
Article in English | MEDLINE | ID: mdl-31091708

ABSTRACT

We performed a three-dimensional (3D) analysis of the microvascular network of the cerebral cortex of twitcher mice (an authentic model of Krabbe disease) using a restricted set of indexes that are able to describe the arrangement of the microvascular tree in CD31-stained sections. We obtained a near-linear graphical "fingerprint" of the microangioarchitecture of wild-type and twitcher animals that describes the amounts, spatial dispersion, and spatial relationships of adjacent classes of caliber-filtered microvessels. We observed significant alterations of the microangioarchitecture of the cerebral cortex of twitcher mice, whereas no alterations occur in renal microvessels, which is keeping with the observation that kidney is an organ that is not affected by the disease. This approach may represent an important starting point for the study of the microvascular changes that occur in the central nervous system (CNS) under different physiopathological conditions.


Subject(s)
Cerebral Cortex/diagnostic imaging , Image Processing, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Leukodystrophy, Globoid Cell/diagnostic imaging , Microvessels/diagnostic imaging , Animals , Cerebral Cortex/blood supply , Mice , Microscopy, Confocal/methods
10.
Int J Mol Sci ; 21(1)2019 Dec 30.
Article in English | MEDLINE | ID: mdl-31905906

ABSTRACT

Krabbe disease (KD) is an autosomal recessive sphingolipidosis caused by the deficiency of the lysosomal hydrolase ß-galactosylceramidase (GALC). Oligodendroglia degeneration and demyelination of the nervous system lead to neurological dysfunctions which are usually lethal by two years of age. At present, the only clinical treatment with any proven efficacy is hematopoietic stem-cell transplantation, which is more effective when administered in the neonatal period to presymptomatic recipients. Bone marrow (BM) sinusoidal endothelial cells (SECs) play a pivotal role in stem cell engraftment and reconstitution of hematopoiesis. Previous observations had shown significant alterations of microvascular endothelial cells in the brain of KD patients and in Galc mutant twitcher mice, an authentic model of the disease. In the present study, we investigated the vascular component of the BM in the femurs of symptomatic homozygous twitcher mice at postnatal day P36. Histological, immunohistochemical, and two-photon microscopy imaging analyses revealed the presence of significant alterations of the diaphyseal BM vasculature, characterized by enlarged, discontinuous, and hemorrhagic SECs that express the endothelial marker vascular endothelial growth factor receptor-2 (VEGFR2) but lack platelet/endothelial cell adhesion molecule-1 (CD31) expression. In addition, computer-aided image analysis indicates that twitcher CD31-/VEGFR2+ SECs show a significant increase in lumen size and in the number and size of endothelial gaps compared to BM SECs of wild type littermates. These results suggest that morphofunctional defects in the BM vascular niche may contribute to the limited therapeutic efficacy of hematopoietic stem-cell transplantation in KD patients at symptomatic stages of the disease.


Subject(s)
Bone Marrow/metabolism , Galactosylceramidase/metabolism , Leukodystrophy, Globoid Cell/metabolism , Animals , Bone Marrow/pathology , Brain/metabolism , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Galactosylceramidase/genetics , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Leukodystrophy, Globoid Cell/genetics , Leukodystrophy, Globoid Cell/pathology , Mice , Mice, Inbred C57BL , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
11.
Int J Mol Sci ; 20(18)2019 Sep 17.
Article in English | MEDLINE | ID: mdl-31533326

ABSTRACT

Fibrosarcoma is an aggressive subtype of soft tissue sarcoma categorized in infantile/congenital-type and adult-type. Fibrosarcoma cells and its surrounding immune inflammatory infiltrates overexpress or induce the expression of fibroblast growth factor-2 (FGF-2) that have a crucial role in tumor progression and angiogenesis. The inflammation-associated long pentraxin 3 (PTX3) was found to reduce FGF-2-mediated angiogenesis, but its role on fibrosarcoma immune inflammatory infiltrate is still unknown. In this study, we have evaluated the PTX3 activity on immune infiltrating mast cells, macrophages and T-lymphocytes by immunohistochemistry on murine MC-TGS17-51 fibrosarcoma cells and on transgenic TgN(Tie2-hPTX3) mouse. In these fibrosarcoma models we found a reduced neovascularization and a significant decrease of inflammatory infiltrate. Indeed, we show that PTX3 reduces the level of complement 3 (C3) deposition reducing fibrosarcoma progression. In conclusion, we hypothesize that targeting fibrosarcoma microenvironment by FGF/FGFR inhibitors may improve treatment outcome.


Subject(s)
C-Reactive Protein/genetics , Fibrosarcoma/etiology , Fibrosarcoma/metabolism , Immunomodulation , Neovascularization, Pathologic/genetics , Serum Amyloid P-Component/genetics , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Animals , C-Reactive Protein/metabolism , Cell Line, Tumor , Disease Models, Animal , Fibrosarcoma/pathology , Gene Expression , Immunohistochemistry , Inflammation/complications , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Male , Mice , Neovascularization, Pathologic/metabolism , Serum Amyloid P-Component/metabolism
12.
Anal Chem ; 90(13): 7855-7861, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29870225

ABSTRACT

Understanding extracellular vesicle (EV) internalization mechanisms and pathways in cells is of capital importance for both EV basic biology and clinical translation, but still presents analytical hurdles, such as undetermined purity grade and/or concentration of the EV samples and lack of standard protocols. We report an accessible, robust, and versatile method for resolving dose-dependent uptake profiles of exosomes-the nanosized (30-150 nm) subtypes of EVs of intracellular origin which are more intensively investigated for diagnostic and therapeutic applications-by cultured cells. The method is based on incubating recipient cells with consistently increasing doses of exosomes which are graded for purity and titrated by a COlorimetric NANoplasmonic (CONAN) assay followed by cell flow cytofluorimetric analysis. The proposed method allowed evaluation and comparison of the uptake of human serum exosomes by cancer cell lines of murine (TRAMP-C2) and human (LNCaP, DU145, MDA-MB-231, and A375) origin, setting a firmer footing for better characterization and understanding of exosome biology in different in vitro and (potentially) in vivo models of cancer growth.


Subject(s)
Exosomes/metabolism , Flow Cytometry/methods , Nanotechnology/methods , Animals , Biological Transport , Cell Line, Tumor , Colloids , Humans , Mice
13.
Pharmacol Res ; 107: 172-185, 2016 05.
Article in English | MEDLINE | ID: mdl-27013279

ABSTRACT

Fibroblast growth factors (FGFs) are a family of pleiotropic factors produced by stromal and parenchymal tumor cells. Even though FGFs have been firstly characterized as angiogenic factors, they exert autocrine and paracrine functions not only on endothelial cells but also on tumor cells and other stromal components. Thus, the FGF/FGF receptor (FGFR) pathway may represent a key player in tumor growth by regulating the complex cross-talk between stromal and tumor compartments. The ligand dependent or independent activation of the FGF/FGFR system by gene upregulation, oncogenic mutation or amplification occurs in a variety of human tumors and is implicated in various key steps of tumor growth and progression. In addition, FGF/FGFR activation has been described as a mechanism of tumor escape in response to antiangiogenic/anti-VEGF therapies. Experimental and clinical evidences provide a compelling biologic rationale for the development of anti-FGF/FGFR targeting agents in cancer therapy. However, the development of drugs specifically targeting the FGF/FGFR pathway proved to be difficult, also due to the high redundancy and pleiotropic effects of FGF and FGFR family members. On the other hand, the possibility to develop "two-compartment" targeting agents endowed with both antiangiogenic and antitumor activities remains promising. Here we will review the preclinical and clinical approaches and potential therapeutics currently available to block the FGF/FGFR system in human cancer.


Subject(s)
Fibroblast Growth Factors/antagonists & inhibitors , Neoplasms/drug therapy , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Animals , Endothelial Cells/metabolism , Fibroblast Growth Factors/metabolism , Humans , Neoplasms/metabolism , Receptors, Fibroblast Growth Factor/metabolism
14.
Angiogenesis ; 18(4): 499-510, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26310512

ABSTRACT

Defects of the angiogenic process occur in the brain of twitcher mouse, an authentic model of human Krabbe disease caused by genetic deficiency of lysosomal ß-galactosylceramidase (GALC), leading to lethal neurological dysfunctions and accumulation of neurotoxic psychosine in the central nervous system. Here, quantitative computational analysis was used to explore the alterations of brain angioarchitecture in twitcher mice. To this aim, customized ImageJ routines were used to assess calibers, amounts, lengths and spatial dispersion of CD31(+) vessels in 3D volumes from the postnatal frontal cortex of twitcher animals. The results showed a decrease in CD31 immunoreactivity in twitcher brain with a marked reduction in total vessel lengths coupled with increased vessel fragmentation. No significant changes were instead observed for the spatial dispersion of brain vessels throughout volumes or in vascular calibers. Notably, no CD31(+) vessel changes were detected in twitcher kidneys in which psychosine accumulates at very low levels, thus confirming the specificity of the effect. Microvascular corrosion casting followed by scanning electron microscopy morphometry confirmed the presence of significant alterations of the functional angioarchitecture of the brain cortex of twitcher mice with reduction in microvascular density, vascular branch remodeling and intussusceptive angiogenesis. Intussusceptive microvascular growth, confirmed by histological analysis, was paralleled by alterations of the expression of intussusception-related genes in twitcher brain. Our data support the hypothesis that a marked decrease in vascular development concurs to the onset of neuropathological lesions in twitcher brain and suggest that neuroinflammation-driven intussusceptive responses may represent an attempt to compensate impaired sprouting angiogenesis.


Subject(s)
Brain/blood supply , Cerebrovascular Circulation , Intussusception/physiopathology , Leukodystrophy, Globoid Cell/physiopathology , Microcirculation , Microvessels/physiopathology , Animals , Disease Models, Animal , Humans , Intussusception/genetics , Intussusception/pathology , Leukodystrophy, Globoid Cell/genetics , Leukodystrophy, Globoid Cell/pathology , Mice
15.
FASEB J ; 28(3): 1132-44, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24265487

ABSTRACT

N6-isopentenyladenosine (iPA), an end product of the mevalonate pathway with an isopentenyl chain, is already known to exert a suppressor effect against various tumors. In this work, we investigated whether iPA also directly interferes with the angiogenic process, which is fundamental to tumor growth and progression. To this end, using human umbilical vein endothelial cells (HUVECs) as a suitable in vitro model of angiogenesis, we evaluated their viability, proliferation, migration, invasion, tube formation in response to iPA, and molecular mechanisms involved. Data were corroborated in mice by using a gel plug assay. iPA dose- and time-dependently inhibited all the neoangiogenesis stages, with an IC50 of 0.98 µM. We demonstrated for the first time, by liquid chromatography-coupled tandem mass spectrometry (LC-MS/MS), that iPA was monophosphorylated into 5'-iPA-monophosphate (iPAMP) by the adenosine kinase (ADK) inside the cells. iPAMP is the active form that inhibits angiogenesis through the direct activation of AMP-kinase (AMPK). Indeed, all effects were completely reversed by pretreatment with 5-iodotubercidin (5-Itu), an ADK inhibitor. The isoprenoid intermediate isopentenyl pyrophosphate (IPP), which shares the isopentenyl moiety with iPA, was ineffective in the inhibition of angiogenesis, thus showing that the iPA structure is specific for the observed effects. In conclusion, iPA is a novel AMPK activator and could represent a useful tool for the treatment of diseases where excessive neoangiogenesis is the underlying pathology.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Angiogenesis Inhibitors/metabolism , Isopentenyladenosine/metabolism , Chromatography, Liquid , Enzyme Activation , Human Umbilical Vein Endothelial Cells , Humans , Neovascularization, Pathologic , Phosphorylation , Tandem Mass Spectrometry
16.
J Pathol ; 230(2): 228-38, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23424081

ABSTRACT

Fibroblast growth factors (FGFs) exert autocrine/paracrine functions in prostate cancer by stimulating angiogenesis and tumour growth. Here dihydrotestosterone (DHT) up-regulates FGF2 and FGF8b production in murine TRAMP-C2 prostate cancer cells, activating a FGF-dependent autocrine loop of stimulation. The soluble pattern recognition receptor long pentraxin-3 (PTX3) acts as a natural FGF antagonist that binds FGF2 and FGF8b via its N-terminal domain. We demonstrate that recombinant PTX3 protein and the PTX3-derived pentapeptide Ac-ARPCA-NH2 abolish the mitogenic response of murine TRAMP-C2 cells and human LNCaP prostate cancer cells to DHT and FGFs. Also, PTX3 hampers the angiogenic activity of DHT-activated TRAMP-C2 cells on the chick embryo chorioallantoic membrane (CAM). Accordingly, human PTX3 overexpression inhibits the mitogenic activity exerted by DHT or FGFs on hPTX3_TRAMP-C2 cell transfectants and their angiogenic activity. Also, hPTX3_TRAMP-C2 cells show a dramatic decrease of their angiogenic and tumourigenic potential when grafted in syngeneic or immunodeficient athymic male mice. A similar inhibitory effect is observed when TRAMP-C2 cells overexpress only the FGF-binding N-terminal PTX3 domain. In keeping with the anti-tumour activity of PTX3 in experimental prostate cancer, immunohistochemical analysis of prostate needle biopsies from primary prostate adenocarcinoma patients shows that parenchymal PTX3 expression, abundant in basal cells of normal glands, is lost in high-grade prostatic intraepithelial neoplasia and in invasive tumour areas. These results identify PTX3 as a potent FGF antagonist endowed with anti-angiogenic and anti-neoplastic activity in prostate cancer.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , C-Reactive Protein/pharmacology , Prostate/drug effects , Prostatic Neoplasms/drug therapy , Serum Amyloid P-Component/pharmacology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Chick Embryo , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/drug effects , Dihydrotestosterone/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mitogens/antagonists & inhibitors , Neovascularization, Physiologic/drug effects , Prostate/metabolism , Prostate/pathology , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Recombinant Proteins/pharmacology
17.
Brain ; 136(Pt 9): 2859-75, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23983033

ABSTRACT

Globoid cell leukodystrophy (Krabbe disease) is a neurological disorder of infants caused by genetic deficiency of the lysosomal enzyme ß-galactosylceramidase leading to accumulation of the neurotoxic metabolite 1-ß-d-galactosylsphingosine (psychosine) in the central nervous system. Angiogenesis plays a pivotal role in the physiology and pathology of the brain. Here, we demonstrate that psychosine has anti-angiogenic properties by causing the disassembling of endothelial cell actin structures at micromolar concentrations as found in the brain of patients with globoid cell leukodystrophy. Accordingly, significant alterations of microvascular endothelium were observed in the post-natal brain of twitcher mice, an authentic model of globoid cell leukodystrophy. Also, twitcher endothelium showed a progressively reduced capacity to respond to pro-angiogenic factors, defect that was corrected after transduction with a lentiviral vector harbouring the murine ß-galactosylceramidase complementary DNA. Finally, RNA interference-mediated ß-galactosylceramidase gene silencing causes psychosine accumulation in human endothelial cells and hampers their mitogenic and motogenic response to vascular endothelial growth factor. Accordingly, significant alterations were observed in human microvasculature from brain biopsy of a globoid cell leukodystrophy case. Together these data demonstrate that ß-galactosylceramidase deficiency induces significant alterations in endothelial neovascular responses that may contribute to central nervous system and systemic damages that occur in globoid cell leukodystrophy.


Subject(s)
Leukodystrophy, Globoid Cell/complications , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/pathology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Aorta/pathology , Aorta/ultrastructure , Biocompatible Materials , Brain/drug effects , Brain/pathology , Brain/ultrastructure , Cattle , Cell Movement/drug effects , Cell Movement/genetics , Chorioallantoic Membrane/drug effects , Chorioallantoic Membrane/metabolism , Collagen/toxicity , Disease Models, Animal , Drug Combinations , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibroblast Growth Factor 2/pharmacology , Green Fluorescent Proteins/metabolism , Humans , Laminin/toxicity , Leukodystrophy, Globoid Cell/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Neovascularization, Pathologic/prevention & control , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Proteoglycans/toxicity , Psychosine/metabolism , Psychosine/pharmacology , RNA, Small Interfering/administration & dosage , Time Factors , Transfection , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Zonula Occludens-1 Protein
18.
Angiogenesis ; 16(3): 707-22, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23605004

ABSTRACT

The proapoptotic death receptor 5 (DR5) expressed by tumor associated endothelial cells (TECs) mediates vascular disrupting effects of human CD34(+) cells engineered to express membrane-bound tumor necrosis factor-related apoptosis-inducing ligand (CD34-TRAIL (+) cells) in mice. Indeed, lack of DR5 on TECs causes resistance to CD34-TRAIL (+) cells. By xenografting in nonobese diabetic/severe combined immunodeficient mice the TRAIL-resistant lymphoma cell line SU-DHL-4V, which generates tumors lacking endothelial DR5 expression, here we demonstrate for the first time that the Akt inhibitor perifosine induces in vivo DR5 expression on TECs, thereby overcoming tumor resistance to the vascular disruption activity of CD34-TRAIL (+) cells. In fact, CD34-TRAIL (+) cells combined with perifosine, but not CD34-TRAIL (+) cells alone, exerted marked antivascular effects and caused a threefold increase of hemorrhagic necrosis in SU-DHL-4V tumors. Consistent with lack of DR5 expression, CD34-TRAIL (+) cells failed to affect the growth of SU-DHL-4V tumors, but CD34-TRAIL (+) cells plus perifosine reduced tumor volumes by 60 % compared with controls. In view of future clinical studies using membrane-bound TRAIL, our results highlight a strategy to rescue patients with primary or acquired resistance due to the lack of DR5 expression in tumor vasculature.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Regulation/drug effects , Neovascularization, Pathologic/physiopathology , Phosphorylcholine/analogs & derivatives , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Analysis of Variance , Animals , Antigens, CD34/metabolism , Blotting, Western , Cell Line, Tumor , DNA Primers/genetics , Female , Flow Cytometry , Fluorescent Antibody Technique , Hematopoietic Stem Cell Transplantation , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Inbred NOD , Mice, SCID , Microscopy, Confocal , Phosphorylcholine/pharmacology , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , TNF-Related Apoptosis-Inducing Ligand/metabolism
19.
Expert Opin Ther Pat ; 33(11): 775-796, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37847492

ABSTRACT

INTRODUCTION: The therapeutic targeting of the ubiquitin-proteasome pathway (UPP) through inhibitors of the 20S proteasome core proteolytic activities has revolutionized the treatment of hematological malignancies and is paving the way for its extension to solid tumors. AREAS COVERED: This review covers the progress made in the field of proteasome inhibitors, ranging from the first-generation bortezomib to the latest second-generation inhibitors such as carfilzomib and ixazomib as well as the proteasome inhibitors in clinical phase such as oprozomib and marizomib. The development of selective and potent proteasome inhibitors with improved pharmacological properties is described from the synthesis to their basic biological, and clinical validation. EXPERT OPINION: Proteasome inhibitors have transformed the treatment landscape for hematological malignancies and hold great promise for cancer therapy. Combination therapies targeting multiple pathways, the development of novel inhibitors or 'hybrid-inhibitors,' and the optimization of treatment protocols are key areas for future exploration. The extension of proteasome inhibitors for the treatment of solid tumors, and their ability to pass the blood-brain barrier open new possibilities for treating central nervous system cancers. However, managing adverse effects, particularly those affecting the central nervous system, remains a critical consideration and a strategic 'working on' aspect for the near future.


Subject(s)
Antineoplastic Agents , Hematologic Neoplasms , Multiple Myeloma , Neoplasms , Humans , Proteasome Inhibitors/adverse effects , Patents as Topic , Antineoplastic Agents/adverse effects , Neoplasms/drug therapy , Hematologic Neoplasms/drug therapy , Multiple Myeloma/drug therapy
20.
Cell Death Discov ; 9(1): 81, 2023 Mar 06.
Article in English | MEDLINE | ID: mdl-36872341

ABSTRACT

Prostate cancer (PCa) is a leading cause of death in the male population commonly treated with androgen deprivation therapy that often relapses as androgen-independent and aggressive castration-resistant prostate cancer (CRPC). Ferroptosis is a recently described form of cell death that requires abundant cytosolic labile iron to promote membrane lipid peroxidation and which can be induced by agents that inhibit the glutathione peroxidase-4 activity such as RSL3. Exploiting in vitro and in vivo human and murine PCa models and the multistage transgenic TRAMP model of PCa we show that RSL3 induces ferroptosis in PCa cells and demonstrate for the first time that iron supplementation significantly increases the effect of RSL3 triggering lipid peroxidation, enhanced intracellular stress and leading to cancer cell death. Moreover, the combination with the second generation anti-androgen drug enzalutamide potentiates the effect of the RSL3 + iron combination leading to superior inhibition of PCa and preventing the onset of CRPC in the TRAMP mouse model. These data open new perspectives in the use of pro-ferroptotic approaches alone or in combination with enzalutamide for the treatment of PCa.

SELECTION OF CITATIONS
SEARCH DETAIL