Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 16(736): eadf9874, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38416843

ABSTRACT

Targeting aromatase deprives ER+ breast cancers of estrogens and is an effective therapeutic approach for these tumors. However, drug resistance is an unmet clinical need. Lipidomic analysis of long-term estrogen-deprived (LTED) ER+ breast cancer cells, a model of aromatase inhibitor resistance, revealed enhanced intracellular lipid storage. Functional metabolic analysis showed that lipid droplets together with peroxisomes, which we showed to be enriched and active in the LTED cells, controlled redox homeostasis and conferred metabolic adaptability to the resistant tumors. This reprogramming was controlled by acetyl-CoA-carboxylase-1 (ACC1), whose targeting selectively impaired LTED survival. However, the addition of branched- and very long-chain fatty acids reverted ACC1 inhibition, a process that was mediated by peroxisome function and redox homeostasis. The therapeutic relevance of these findings was validated in aromatase inhibitor-treated patient-derived samples. Last, targeting ACC1 reduced tumor growth of resistant patient-derived xenografts, thus identifying a targetable hub to combat the acquisition of estrogen independence in ER+ breast cancers.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Aromatase Inhibitors/pharmacology , Aromatase Inhibitors/therapeutic use , Peroxisomes/metabolism , Peroxisomes/pathology , Acetyl-CoA Carboxylase , Lipid Droplets/metabolism , Lipid Droplets/pathology , Cell Line, Tumor , Estrogens/metabolism , Drug Resistance, Neoplasm
2.
Sci Adv ; 9(37): eadh4184, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37713487

ABSTRACT

Cancers feature substantial intratumoral heterogeneity of genetic and phenotypically distinct lineages. Although interactions between coexisting lineages are emerging as a potential contributor to tumor evolution, the extent and nature of these interactions remain largely unknown. We postulated that tumors develop ecological interactions that sustain diversity and facilitate metastasis. Using a combination of fluorescent barcoding, mathematical modeling, metabolic analysis, and in vivo models, we show that the Allee effect, i.e., growth dependency on population size, is a feature of tumor lineages and that cooperative ecological interactions between lineages alleviate the Allee barriers to growth in a model of triple-negative breast cancer. Soluble metabolite exchange formed the basis for these cooperative interactions and catalyzed the establishment of a polyclonal community that displayed enhanced metastatic dissemination and outgrowth in xenograft models. Our results highlight interclonal metabolite exchange as a key modulator of tumor ecology and a contributing factor to overcoming Allee effect-associated growth barriers to metastasis.


Subject(s)
Coloring Agents , Triple Negative Breast Neoplasms , Humans , Animals , Disease Models, Animal , Population Density
3.
Mol Cancer Ther ; 22(6): 790-804, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36921314

ABSTRACT

Next-generation Trop-2-targeted therapy against advanced cancers is hampered by expression of Trop-2 in normal tissues. We discovered that Trop-2 undergoes proteolytic activation by ADAM10 in cancer cells, leading to the exposure of a previously inaccessible protein groove flanked by two N-glycosylation sites. We designed a recognition strategy for this region, to drive selective cancer vulnerability in patients. Most undiscriminating anti-Trop-2 mAbs recognize a single immunodominant epitope. Hence, we removed it by deletion mutagenesis. Cancer-specific, glycosylation-prone mAbs were selected by ELISA, bio-layer interferometry, flow cytometry, confocal microscopy for differential binding to cleaved/activated, wild-type and glycosylation site-mutagenized Trop-2. The resulting 2G10 mAb family binds Trop-2-expressing cancer cells, but not Trop-2 on normal cells. We humanized 2G10 by state-of-the-art complementarity determining region grafting/re-modeling, yielding Hu2G10. This antibody binds cancer-specific, cleaved/activated Trop-2 with Kd < 10-12 mol/L, and uncleaved/wtTrop-2 in normal cells with Kd 3.16×10-8 mol/L, thus promising an unprecedented therapeutic index in patients. In vivo, Hu2G10 ablates growth of Trop-2-expressing breast, colon, prostate cancers, but shows no evidence of systemic toxicity, paving the way for a paradigm shift in Trop-2-targeted therapy.


Subject(s)
Immunoconjugates , Prostatic Neoplasms , Male , Humans , Antigens, Neoplasm/genetics , Antibodies, Monoclonal/pharmacology
5.
Nat Mater ; 22(5): 644-655, 2023 05.
Article in English | MEDLINE | ID: mdl-36581770

ABSTRACT

The process in which locally confined epithelial malignancies progressively evolve into invasive cancers is often promoted by unjamming, a phase transition from a solid-like to a liquid-like state, which occurs in various tissues. Whether this tissue-level mechanical transition impacts phenotypes during carcinoma progression remains unclear. Here we report that the large fluctuations in cell density that accompany unjamming result in repeated mechanical deformations of cells and nuclei. This triggers a cellular mechano-protective mechanism involving an increase in nuclear size and rigidity, heterochromatin redistribution and remodelling of the perinuclear actin architecture into actin rings. The chronic strains and stresses associated with unjamming together with the reduction of Lamin B1 levels eventually result in DNA damage and nuclear envelope ruptures, with the release of cytosolic DNA that activates a cGAS-STING (cyclic GMP-AMP synthase-signalling adaptor stimulator of interferon genes)-dependent cytosolic DNA response gene program. This mechanically driven transcriptional rewiring ultimately alters the cell state, with the emergence of malignant traits, including epithelial-to-mesenchymal plasticity phenotypes and chemoresistance in invasive breast carcinoma.


Subject(s)
Actins , Neoplasms , DNA , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Cytosol/metabolism , Signal Transduction
7.
Small ; 18(17): e2106097, 2022 04.
Article in English | MEDLINE | ID: mdl-35344274

ABSTRACT

Circulating tumor cell (CTC) clusters are associated with increased metastatic potential and worse patient prognosis, but are rare, difficult to count, and poorly characterized biophysically. The PillarX device described here is a bimodular microfluidic device (Pillar-device and an X-magnetic device) to profile single CTCs and clusters from whole blood based on their size, deformability, and epithelial marker expression. Larger, less deformable clusters and large single cells are captured in the Pillar-device and sorted according to pillar gap sizes. Smaller, deformable clusters and single cells are subsequently captured in the X-device and separated based on epithelial marker expression using functionalized magnetic nanoparticles. Clusters of established and primary breast cancer cells with variable degrees of cohesion driven by different cell-cell adhesion protein expression are profiled in the device. Cohesive clusters exhibit a lower deformability as they travel through the pillar array, relative to less cohesive clusters, and have greater collective invasive behavior. The ability of the PillarX device to capture clusters is validated in mouse models and patients of metastatic breast cancer. Thus, this device effectively enumerates and profiles CTC clusters based on their unique geometrical, physical, and biochemical properties, and could form the basis of a novel prognostic clinical tool.


Subject(s)
Breast Neoplasms , Neoplastic Cells, Circulating , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Separation , Female , Humans , Lab-On-A-Chip Devices , Mice , Neoplastic Cells, Circulating/pathology , Prognosis
8.
Oncogene ; 41(12): 1795-1808, 2022 03.
Article in English | MEDLINE | ID: mdl-35132180

ABSTRACT

Trop-2 is a transmembrane signal transducer that is overexpressed in most human cancers, and drives malignant progression. To gain knowledge on the higher-order molecular mechanisms that drive Trop-2 signaling, we applied next-generation sequencing, proteomics, and high-resolution microscopy to models and primary cases of human colorectal cancer (CRC). We had previously shown that Trop-2 induces a Ca2+ signal. We reveal here that Trop-2 binds the cell membrane Na+/K+-ATPase, and that clustering of Trop-2 induces an intracellular Ca2+ rise followed by membrane translocation of PKCα, which in turn phosphorylates the Trop-2 cytoplasmic tail. This feed-forward signaling is promoted by the binding of Trop-2 to the PKCα membrane-anchor CD9. CRISPR-based inactivation of CD9 in CRC cells shows that CD9 is required by Trop-2 for recruiting PKCα and cofilin-1 to the cell membrane. This induces malignant progression through proteolytic cleavage of E-cadherin, remodeling of the ß-actin cytoskeleton, and activation of Akt and ERK. The interaction between Trop-2 and CD9 was validated in vivo in murine models of CRC growth and invasion. Overexpression of the components of this Trop-2-driven super-complex significantly worsened disease-free and overall survival of CRC patients, supporting a pivotal relevance in CRC malignant progression. Our findings demonstrate a previously unsuspected layer of cancer growth regulation, which is dormant in normal tissues, and is activated by Trop-2 in cancer cells.


Subject(s)
Colorectal Neoplasms , Protein Kinase C-alpha , Actin Depolymerizing Factors/metabolism , Adenosine Triphosphatases/metabolism , Animals , Colorectal Neoplasms/pathology , Humans , Mice , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Signal Transduction , Tetraspanin 29
9.
Nat Commun ; 11(1): 4828, 2020 09 24.
Article in English | MEDLINE | ID: mdl-32973141

ABSTRACT

ATR responds to mechanical stress at the nuclear envelope and mediates envelope-associated repair of aberrant topological DNA states. By combining microscopy, electron microscopic analysis, biophysical and in vivo models, we report that ATR-defective cells exhibit altered nuclear plasticity and YAP delocalization. When subjected to mechanical stress or undergoing interstitial migration, ATR-defective nuclei collapse accumulating nuclear envelope ruptures and perinuclear cGAS, which indicate loss of nuclear envelope integrity, and aberrant perinuclear chromatin status. ATR-defective cells also are defective in neuronal migration during development and in metastatic dissemination from circulating tumor cells. Our findings indicate that ATR ensures mechanical coupling of the cytoskeleton to the nuclear envelope and accompanying regulation of envelope-chromosome association. Thus the repertoire of ATR-regulated biological processes extends well beyond its canonical role in triggering biochemical implementation of the DNA damage response.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Nucleus/metabolism , Stress, Mechanical , Actin Cytoskeleton , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Brain , Chromatin , Cytoplasm , Cytoskeleton/metabolism , DNA Damage , Mice, Knockout , Neoplasm Metastasis , Neurogenesis , Nuclear Envelope/metabolism
10.
J Clin Med ; 9(1)2019 Dec 29.
Article in English | MEDLINE | ID: mdl-31905780

ABSTRACT

Tumor recurrence is now the leading cause of breast cancer-related death. These recurrences are believed to arise from residual cancer stem cells that survive initial therapeutic intervention. Therefore, a comprehensive understanding of cancer stem cell biology is needed to generate more effective therapies. Here we investigate the association between dysregulation of lipid metabolism and breast cancer stem cells. Focusing specifically on lipid droplets, we found that the lipid droplet number correlates with stemness in a panel of breast cell lines. Using a flow cytometry-based method developed for this study, we establish a means to isolate cells with augmented lipid droplet loads from total populations and show that they are enriched in cancer stem cells. Furthermore, pharmacological targeting of fatty acid metabolism reveals a metabolic addiction in a subset of cell lines. Our results highlight a key role for the lipid metabolism in the maintenance of the breast cancer stem cell pool, and as such, suggest it as a potential therapeutic target.

11.
J Clin Invest ; 127(6): 2091-2105, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28504653

ABSTRACT

Tumor recurrence is the leading cause of breast cancer-related death. Recurrences are largely driven by cancer cells that survive therapeutic intervention. This poorly understood population is referred to as minimal residual disease. Here, using mouse models that faithfully recapitulate human disease together with organoid cultures, we have demonstrated that residual cells acquire a transcriptionally distinct state from normal epithelium and primary tumors. Gene expression changes and functional characterization revealed altered lipid metabolism and elevated ROS as hallmarks of the cells that survive tumor regression. These residual cells exhibited increased oxidative DNA damage, potentiating the acquisition of somatic mutations during hormonal-induced expansion of the mammary cell population. Inhibition of either cellular fatty acid synthesis or fatty acid transport into mitochondria reduced cellular ROS levels and DNA damage, linking these features to lipid metabolism. Direct perturbation of these hallmarks in vivo, either by scavenging ROS or by halting the cyclic mammary cell population expansion, attenuated tumor recurrence. Finally, these observations were mirrored in transcriptomic and histological signatures of residual cancer cells from neoadjuvant-treated breast cancer patients. These results highlight the potential of lipid metabolism and ROS as therapeutic targets for reducing tumor recurrence in breast cancer patients.


Subject(s)
Breast Neoplasms/metabolism , Neoplasm Recurrence, Local/metabolism , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Lapatinib , Lipid Metabolism , Metabolic Networks and Pathways , Mice , Neoplasm Recurrence, Local/prevention & control , Neoplasm, Residual , Oxidative Stress , Progesterone/pharmacology , Quinazolines/pharmacology , Reactive Oxygen Species/metabolism , Transcriptome , Xenograft Model Antitumor Assays
12.
Curr Gene Ther ; 14(3): 161-9, 2014.
Article in English | MEDLINE | ID: mdl-24828254

ABSTRACT

BACKGROUND: DNA vaccines provide high tolerability and safety but commonly suffer from suboptimal immunogenicity. We previously reported that a plasmid vector (pATRex), encoding the DNA sequence for the von Willebrand I/A domain of the tumor endothelial marker-8 (TEM8) when given in combination with plasmid-encoded tumor antigens acted as a powerful molecular adjuvant enhancing immunity against breast and melanoma tumors. AIMS: In the present study we addressed two unsolved issues; would the adjuvant action of pATRex extend to a DNA vaccine against infectious disease and, if so, what is the mechanistic basis for pATRex adjuvant action? RESULTS: Here we show in a murine malaria vaccine model that co-administration of pATRex potentiates antibody production elicited by an intramuscular injection of plasmid encoding Plasmodium yoelii merozoite surface protein 4/5 (PyMSP4/5). pATRex enhanced the B-cell response and induced increased IgG1 production consistent with TH2 polarization of the DNA vaccine response. To explore the mechanism of adjuvant action, cells were transfected in vitro with pATRex and this resulted in formation of insoluble intracellular aggregates and apoptotic cell death. Using a structural modeling approach we identified a short peptide sequence (α3-ß4) within ATRex responsible for protein aggregation and confirmed that transfection of cells with plasmid encoding this self-assembling peptide similarly triggered intracellular aggregates, caspase activation and cell death. CONCLUSION: Plasmids encoding aggregation-promoting domains induce formation of insoluble intracellular aggregates that trigger caspase activation and apoptotic cell death leading to activation of the innate immune system thereby acting as genetic adjuvants.


Subject(s)
Adjuvants, Immunologic/genetics , Malaria Vaccines/immunology , Plasmids/genetics , Vaccines, DNA/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , B-Lymphocytes/immunology , Disease Models, Animal , Female , Malaria/therapy , Mice , Mice, Inbred BALB C , Plasmodium yoelii , Protein Aggregates/genetics , Protein Aggregates/immunology
13.
Mol Cell Biol ; 29(15): 4057-66, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19470759

ABSTRACT

Regulation of the Saccharomyces cerevisiae HO promoter has been shown to require the recruitment of chromatin-modifying and -remodeling enzymes. Despite this, relatively little is known about what changes to chromatin structure occur during the course of regulation at HO. Here, we used indirect end labeling in synchronized cultures to show that the chromatin structure is disrupted in a region that spans bp -600 to -1800 relative to the transcriptional start site. Across this region, there is a loss of canonical nucleosomes and a reduction in histone DNA cross-linking, as monitored by chromatin immunoprecipitation. The ATPase Snf2 is required for these alterations, but the histone acetyltransferase Gcn5 is not. This suggests that the SWI/SNF complex is directly involved in nucleosome removal at HO. We also present evidence indicating that the histone chaperone Asf1 assists in this. These observations suggest that SWI/SNF-related complexes in concert with histone chaperones act to remove histone octamers from DNA during the course of gene regulation.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Histones/metabolism , Molecular Chaperones/metabolism , Promoter Regions, Genetic/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Cell Cycle Proteins/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/genetics , Deoxyribonucleases, Type II Site-Specific/genetics , Gene Expression Regulation, Fungal , Molecular Chaperones/genetics , Mutation , Nucleosomes/metabolism , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics
14.
Mol Biol Cell ; 13(6): 2106-19, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12058073

ABSTRACT

Nuclear receptors, including the androgen receptor (AR), regulate target cell transcription through interaction with auxiliary proteins to modify chromatin structure. We describe herein a novel AR-interacting protein, termed ARIP4, that has structural features typical of the SNF2-like protein family. With regard to the Snf2 domain, the closest homolog of ARIP4 is the ATRX protein. ARIP4 is a nuclear protein and comprises 1466 amino acids. It interacts with AR in vitro and in cultured yeast and mammalian cells. ARIP4 can be labeled with 8-azido-[gamma-32P]ATP and exhibits DNA-dependent ATPase activity. Like several ATP-dependent chromatin remodeling proteins, ARIP4 generates superhelical torsion within linear DNA fragments in an ATP-dependent manner. With a stably integrated target promoter, ARIP4 elicits a modest enhancement of AR-dependent transactivation. In transient cotransfection assays, ARIP4 modulates AR function in a promoter-dependent manner; it enhances receptor activity on minimal promoters, but does not activate more complex promoters. ARIP4 mutants devoid of ATPase activity fail to alter DNA topology and behave as trans-dominant negative regulators of AR function in transient assays.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Nuclear Proteins , Receptors, Androgen/metabolism , Saccharomyces cerevisiae/physiology , Transcription Factors/metabolism , Transcription, Genetic , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins/genetics , COS Cells , Chlorocebus aethiops , DNA, Complementary , Genes, Reporter , Insecta , Molecular Sequence Data , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins , Sequence Alignment , Serine Endopeptidases/genetics , Transfection , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...