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










Publication year range
1.
Cell Death Discov ; 10(1): 12, 2024 Jan 06.
Article in English | MEDLINE | ID: mdl-38184644

ABSTRACT

Mammary gland development occurs primarily in adulthood, undergoing extensive expansion during puberty followed by cycles of functional specialization and regression with every round of pregnancy/lactation/involution. This process is ultimately driven by the coordinated proliferation and differentiation of mammary epithelial cells. However, the endogenous molecular factors regulating these developmental dynamics are still poorly defined. Endocannabinoid signaling is known to determine cell fate-related events during the development of different organs in the central nervous system and the periphery. Here, we report that the endocannabinoid-degrading enzyme fatty acid amide hydrolase (FAAH) plays a pivotal role in adult mammary gland development. Specifically, it is required for luminal lineage specification in the mammary gland, and it promotes hormone-driven secretory differentiation of mammary epithelial cells by controlling the endogenous levels of anandamide and the subsequent activation of cannabinoid CB1 receptors. Together, our findings shed light on the role of the endocannabinoid system in breast development and point to FAAH as a therapeutic target in milk-production deficits.

2.
Nat Commun ; 14(1): 3130, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37253733

ABSTRACT

Clinical management of breast cancer (BC) metastasis remains an unmet need as it accounts for 90% of BC-associated mortality. Although the luminal subtype, which represents >70% of BC cases, is generally associated with a favorable outcome, it is susceptible to metastatic relapse as late as 15 years after treatment discontinuation. Seeking therapeutic approaches as well as screening tools to properly identify those patients with a higher risk of recurrence is therefore essential. Here, we report that the lipid-degrading enzyme fatty acid amide hydrolase (FAAH) is a predictor of long-term survival in patients with luminal BC, and that it blocks tumor progression and lung metastasis in cell and mouse models of BC. Together, our findings highlight the potential of FAAH as a biomarker with prognostic value in luminal BC and as a therapeutic target in metastatic disease.


Subject(s)
Amidohydrolases , Biomarkers, Tumor , Lung Neoplasms , Animals , Mice , Amidohydrolases/genetics , Lung Neoplasms/pathology , Neoplasm Recurrence, Local/pathology
3.
Cancers (Basel) ; 15(9)2023 Apr 24.
Article in English | MEDLINE | ID: mdl-37173906

ABSTRACT

Melanoma is one of the deadliest forms of cancer. Most melanoma deaths are caused by distant metastases in several organs, especially the brain, the so-called melanoma brain metastases (MBMs). However, the precise mechanisms that sustain the growth of MBMs remain elusive. Recently, the excitatory neurotransmitter glutamate has been proposed as a brain-specific, pro-tumorigenic signal for various types of cancers, but how neuronal glutamate shuttling onto metastases is regulated remains unknown. Here, we show that the cannabinoid CB1 receptor (CB1R), a master regulator of glutamate output from nerve terminals, controls MBM proliferation. First, in silico transcriptomic analysis of cancer-genome atlases indicated an aberrant expression of glutamate receptors in human metastatic melanoma samples. Second, in vitro experiments conducted on three different melanoma cell lines showed that the selective blockade of glutamatergic NMDA receptors, but not AMPA or metabotropic receptors, reduces cell proliferation. Third, in vivo grafting of melanoma cells in the brain of mice selectively devoid of CB1Rs in glutamatergic neurons increased tumour cell proliferation in concert with NMDA receptor activation, whereas melanoma cell growth in other tissue locations was not affected. Taken together, our findings demonstrate an unprecedented regulatory role of neuronal CB1Rs in the MBM tumour microenvironment.

4.
Proc Natl Acad Sci U S A ; 116(9): 3863-3872, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30733293

ABSTRACT

Although human epidermal growth factor receptor 2 (HER2)-targeted therapies have dramatically improved the clinical outcome of HER2-positive breast cancer patients, innate and acquired resistance remains an important clinical challenge. New therapeutic approaches and diagnostic tools for identification, stratification, and treatment of patients at higher risk of resistance and recurrence are therefore warranted. Here, we unveil a mechanism controlling the oncogenic activity of HER2: heteromerization with the cannabinoid receptor CB2R. We show that HER2 physically interacts with CB2R in breast cancer cells, and that the expression of these heteromers correlates with poor patient prognosis. The cannabinoid Δ9-tetrahydrocannabinol (THC) disrupts HER2-CB2R complexes by selectively binding to CB2R, which leads to (i) the inactivation of HER2 through disruption of HER2-HER2 homodimers, and (ii) the subsequent degradation of HER2 by the proteasome via the E3 ligase c-CBL. This in turn triggers antitumor responses in vitro and in vivo. Selective targeting of CB2R transmembrane region 5 mimicked THC effects. Together, these findings define HER2-CB2R heteromers as new potential targets for antitumor therapies and biomarkers with prognostic value in HER2-positive breast cancer.


Subject(s)
Breast Neoplasms/cerebrospinal fluid , Molecular Targeted Therapy , Receptor, Cannabinoid, CB2/genetics , Receptor, ErbB-2/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Dronabinol/pharmacology , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Protein Multimerization/drug effects , Proto-Oncogene Proteins c-cbl/genetics , Receptor, Cannabinoid, CB2/chemistry , Receptor, ErbB-2/chemistry , Signal Transduction
5.
Biochem Pharmacol ; 157: 285-293, 2018 11.
Article in English | MEDLINE | ID: mdl-29940172

ABSTRACT

Breast cancer is the second leading cause of death among women. Although early diagnosis and development of new treatments have improved their prognosis, many patients present innate or acquired resistance to current therapies. New therapeutic approaches are therefore warranted for the management of this disease. Extensive preclinical research has demonstrated that cannabinoids, the active ingredients of Cannabis sativa, trigger antitumor responses in different models of cancer. Most of these studies have been conducted with pure compounds, mainly Δ9-tetrahydrocannabinol (THC). The cannabis plant, however, produces hundreds of other compounds with their own therapeutic potential and the capability to induce synergic responses when combined, the so-called "entourage effect". Here, we compared the antitumor efficacy of pure THC with that of a botanical drug preparation (BDP). The BDP was more potent than pure THC in producing antitumor responses in cell culture and animal models of ER+/PR+, HER2+ and triple-negative breast cancer. This increased potency was not due to the presence of the 5 most abundant terpenes in the preparation. While pure THC acted by activating cannabinoid CB2 receptors and generating reactive oxygen species, the BDP modulated different targets and mechanisms of action. The combination of cannabinoids with estrogen receptor- or HER2-targeted therapies (tamoxifen and lapatinib, respectively) or with cisplatin, produced additive antiproliferative responses in cell cultures. Combinations of these treatments in vivo showed no interactions, either positive or negative. Together, our results suggest that standardized cannabis drug preparations, rather than pure cannabinoids, could be considered as part of the therapeutic armamentarium to manage breast cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Cannabis , Dronabinol/therapeutic use , Phytotherapy , Animals , Cell Line, Tumor , Female , Humans , Mice, Nude , Plant Preparations/therapeutic use , Receptor, ErbB-2/analysis , Triple Negative Breast Neoplasms/drug therapy
6.
Oncotarget ; 7(30): 47565-47575, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27340777

ABSTRACT

The orphan G protein-coupled receptor GPR55 has been directly or indirectly related to basic alterations that drive malignant growth: uncontrolled cancer cell proliferation, sustained angiogenesis, and cancer cell adhesion and migration. However, little is known about the involvement of this receptor in metastasis. Here, we show that elevated GPR55 expression in human tumors is associated with the aggressive basal/triple-negative breast cancer population, higher probability to develop metastases, and therefore poor patient prognosis. Activation of GPR55 by its proposed endogenous ligand lysophosphatidylinositol confers pro-invasive features on breast cancer cells both in vitro and in vivo. Specifically, this effect is elicited by coupling to Gq/11 heterotrimeric proteins and the subsequent activation, through ERK, of the transcription factor ETV4/PEA3. Together, these data show that GPR55 promotes breast cancer metastasis, and supports the notion that this orphan receptor may constitute a new therapeutic target and potential biomarker in the highly aggressive triple-negative subtype.


Subject(s)
Lysophospholipids/pharmacology , Receptors, G-Protein-Coupled/physiology , Triple Negative Breast Neoplasms/pathology , Adenovirus E1A Proteins/physiology , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/physiology , Female , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Humans , Neoplasm Metastasis , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-ets , Receptors, Cannabinoid , rhoA GTP-Binding Protein/physiology
7.
J Med Chem ; 58(9): 3757-66, 2015 May 14.
Article in English | MEDLINE | ID: mdl-25906180

ABSTRACT

Angiogenesis is a requirement for the sustained growth and proliferation of solid tumors, and the development of new compounds that induce a sustained inhibition of the proangiogenic signaling generated by tumor hypoxia still remains as an important unmet need. In this work, we describe a new antiangiogenic compound (22) that inhibits proangiogenic signaling under hypoxic conditions in breast cancer cells. Compound 22 blocks the MAPK pathway, impairs cellular migration under hypoxic conditions, and regulates a set of genes related to angiogenesis. These responses are mediated by HIF-1α, since the effects of compound 22 mostly disappear when its expression is knocked-down. Furthermore, administration of compound 22 in a xenograft model of breast cancer produced tumor growth reductions ranging from 46 to 55% in 38% of the treated animals without causing any toxic side effects. Importantly, in the responding tumors, a significant reduction in the number of blood vessels was observed, further supporting the mechanism of action of the compound. These findings provide a rationale for the development of new antiangiogenic compounds that could eventually lead to new drugs suitable for the treatment of some types of tumors either alone or in combination with other agents.


Subject(s)
Angiogenesis Inhibitors/chemistry , Benzamides/chemistry , Carbamates/chemistry , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/pharmacology , Animals , Benzamides/chemical synthesis , Benzamides/pharmacology , Breast Neoplasms/blood supply , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carbamates/chemical synthesis , Carbamates/pharmacology , Cell Hypoxia , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Signal Transduction , Structure-Activity Relationship
8.
J Natl Cancer Inst ; 107(6): djv077, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25855725

ABSTRACT

BACKGROUND: Pharmacological activation of cannabinoid receptors elicits antitumoral responses in different cancer models. However, the biological role of these receptors in tumor physio-pathology is still unknown. METHODS: We analyzed CB2 cannabinoid receptor protein expression in two series of 166 and 483 breast tumor samples operated in the University Hospitals of Kiel, Tübingen, and Freiburg between 1997 and 2010 and CB2 mRNA expression in previously published DNA microarray datasets. The role of CB2 in oncogenesis was studied by generating a mouse line that expresses the human V-Erb-B2 Avian Erythroblastic Leukemia Viral Oncogene Homolog 2 (HER2) rat ortholog (neu) and lacks CB2 and by a variety of biochemical and cell biology approaches in human breast cancer cells in culture and in vivo, upon modulation of CB2 expression by si/shRNAs and overexpression plasmids. CB2-HER2 molecular interaction was studied by colocalization, coimmunoprecipitation, and proximity ligation assays. Statistical tests were two-sided. RESULTS: We show an association between elevated CB2 expression in HER2+ breast tumors and poor patient prognosis (decreased overall survival, hazard ratio [HR] = 0.29, 95% confidence interval [CI] = 0.09 to 0.71, P = .009) and higher probability to suffer local recurrence (HR = 0.09, 95% CI = 0.049 to 0.54, P = .003) and to develop distant metastases (HR = 0.33, 95% CI = 0.13 to 0.75, P = .009). We also demonstrate that genetic inactivation of CB2 impairs tumor generation and progression in MMTV-neu mice. Moreover, we show that HER2 upregulates CB2 expression by activating the transcription factor ELK1 via the ERK cascade and that an increased CB2 expression activates the HER2 pro-oncogenic signaling at the level of the tyrosine kinase c-SRC. Finally, we show HER2 and CB2 form heteromers in cancer cells. CONCLUSIONS: Our findings reveal an unprecedented role of CB2 as a pivotal regulator of HER2 pro-oncogenic signaling in breast cancer, and they suggest that CB2 may be a biomarker with prognostic value in these tumors.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Receptor, Cannabinoid, CB2/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Germany , Humans , Immunohistochemistry , Immunoprecipitation , Kaplan-Meier Estimate , Mice , Prognosis , RNA, Messenger/metabolism , Receptor, Cannabinoid, CB2/genetics , Receptor, ErbB-2/genetics , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Tissue Array Analysis , Transcription, Genetic
9.
J Med Chem ; 58(5): 2256-64, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25671648

ABSTRACT

Triple-negative breast cancer (TNBC) represents a subtype of breast cancer characterized by high aggressiveness. There is no current targeted therapy for these patients whose prognosis, as a group, is very poor. Here, we report the synthesis and evaluation of a potent antitumor agent in vivo for this type of breast cancer designed as a combination of quinone/cannabinoid pharmacophores. This new compound (10) has been selected from a series of chromenopyrazolediones with full selectivity for the nonpsychotropic CB2 cannabinoid receptor and with efficacy in inducing death of human TNBC cell lines. The dual concept quinone/cannabinoid was supported by the fact that compound 10 exerts antitumor effect by inducing cell apoptosis through activation of CB2 receptors and through oxidative stress. Notably, it did not show either cytotoxicity on noncancerous human mammary epithelial cells nor toxic effects in vivo, suggesting that it may be a new therapeutic tool for the management of TNBC.


Subject(s)
Antineoplastic Agents/pharmacology , Benzopyrans/pharmacology , Benzoquinones/chemistry , Breast/drug effects , Cannabinoids/chemistry , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB2/metabolism , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Benzopyrans/chemistry , Blotting, Western , Breast/cytology , Cell Proliferation/drug effects , Cells, Cultured , Female , Humans , Mice , Mice, Nude , Pyrazoles/chemistry , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, Cannabinoid, CB2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
10.
J Biol Chem ; 289(32): 21960-72, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-24942731

ABSTRACT

The G protein-coupled receptors CB2 (CB2R) and GPR55 are overexpressed in cancer cells and human tumors. Because a modulation of GPR55 activity by cannabinoids has been suggested, we analyzed whether this receptor participates in cannabinoid effects on cancer cells. Here we show that CB2R and GPR55 form heteromers in cancer cells, that these structures possess unique signaling properties, and that modulation of these heteromers can modify the antitumoral activity of cannabinoids in vivo. These findings unveil the existence of previously unknown signaling platforms that help explain the complex behavior of cannabinoids and may constitute new targets for therapeutic intervention in oncology.


Subject(s)
Neoplasms/metabolism , Receptor, Cannabinoid, CB2/chemistry , Receptor, Cannabinoid, CB2/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cannabinoids/metabolism , Cannabinoids/pharmacology , Cell Line, Tumor , Dronabinol/pharmacology , Female , Gene Targeting , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Nude , Neoplasms/drug therapy , Neoplasms/genetics , Protein Structure, Quaternary , RNA, Small Interfering/genetics , Receptor, Cannabinoid, CB2/genetics , Receptors, Cannabinoid , Receptors, G-Protein-Coupled/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...