Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Cell Biol ; 25(12): 1821-1832, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38049604

ABSTRACT

Lineage transitions are a central feature of prostate development, tumourigenesis and treatment resistance. While epigenetic changes are well known to drive prostate lineage transitions, it remains unclear how upstream metabolic signalling contributes to the regulation of prostate epithelial identity. To fill this gap, we developed an approach to perform metabolomics on primary prostate epithelial cells. Using this approach, we discovered that the basal and luminal cells of the prostate exhibit distinct metabolomes and nutrient utilization patterns. Furthermore, basal-to-luminal differentiation is accompanied by increased pyruvate oxidation. We establish the mitochondrial pyruvate carrier and subsequent lactate accumulation as regulators of prostate luminal identity. Inhibition of the mitochondrial pyruvate carrier or supplementation with exogenous lactate results in large-scale chromatin remodelling, influencing both lineage-specific transcription factors and response to antiandrogen treatment. These results establish reciprocal regulation of metabolism and prostate epithelial lineage identity.


Subject(s)
Monocarboxylic Acid Transporters , Prostate , Male , Humans , Prostate/metabolism , Monocarboxylic Acid Transporters/metabolism , Cell Differentiation/physiology , Epithelial Cells/metabolism , Androgen Antagonists/pharmacology , Androgen Antagonists/metabolism , Lactates/metabolism
2.
Sci Signal ; 16(810): eabo5213, 2023 11 07.
Article in English | MEDLINE | ID: mdl-37934809

ABSTRACT

Dysregulated Notch signaling is a common feature of cancer; however, its effects on tumor initiation and progression are highly variable, with Notch having either oncogenic or tumor-suppressive functions in various cancers. To better understand the mechanisms that regulate Notch function in cancer, we studied Notch signaling in a Drosophila tumor model, prostate cancer-derived cell lines, and tissue samples from patients with advanced prostate cancer. We demonstrated that increased activity of the Src-JNK pathway in tumors inactivated Notch signaling because of JNK pathway-mediated inhibition of the expression of the gene encoding the Notch S2 cleavage protease, Kuzbanian, which is critical for Notch activity. Consequently, inactive Notch accumulated in cells, where it was unable to transcribe genes encoding its target proteins, many of which have tumor-suppressive activities. These findings suggest that Src-JNK activity in tumors predicts Notch activity status and that suppressing Src-JNK signaling could restore Notch function in tumors, offering opportunities for diagnosis and targeted therapies for a subset of patients with advanced prostate cancer.


Subject(s)
Drosophila Proteins , Prostatic Neoplasms , Animals , Male , Humans , Drosophila Proteins/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Drosophila , Signal Transduction , Prostatic Neoplasms/metabolism
3.
Cell Rep ; 42(10): 113221, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37815914

ABSTRACT

Advanced prostate cancers are treated with therapies targeting the androgen receptor (AR) signaling pathway. While many tumors initially respond to AR inhibition, nearly all develop resistance. It is critical to understand how prostate tumor cells respond to AR inhibition in order to exploit therapy-induced phenotypes prior to the outgrowth of treatment-resistant disease. Here, we comprehensively characterize the effects of AR blockade on prostate cancer metabolism using transcriptomics, metabolomics, and bioenergetics approaches. The metabolic response to AR inhibition is defined by reduced glycolysis, robust elongation of mitochondria, and increased reliance on mitochondrial oxidative metabolism. We establish DRP1 activity and MYC signaling as mediators of AR-blockade-induced metabolic phenotypes. Rescuing DRP1 phosphorylation after AR inhibition restores mitochondrial fission, while rescuing MYC restores glycolytic activity and prevents sensitivity to complex I inhibition. Our study provides insight into the regulation of treatment-induced metabolic phenotypes and vulnerabilities in prostate cancer.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Prostatic Neoplasms , Humans , Male , Androgens/metabolism , Cell Line, Tumor , Prostatic Neoplasms/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Androgen/drug effects , Receptors, Androgen/metabolism , Signal Transduction
4.
Biotechnol J ; 18(6): e2200434, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36905340

ABSTRACT

3D cancer spheroids represent a highly promising model for study of cancer progression and therapeutic development. Wide-scale adoption of cancer spheroids, however, remains a challenge due to the lack of control over hypoxic gradients that may cloud the assessment of cell morphology and drug response. Here, we present a Microwell Flow Device (MFD) that generates in-well laminar flow around 3D tissues via repetitive tissue sedimentation. Using a prostate cancer cell line, we demonstrate the spheroids in the MFD exhibit improved cell growth, reduced necrotic core formation, enhanced structural integrity, and downregulated expression of cell stress genes. The flow-cultured spheroids also exhibit an improved sensitivity to chemotherapy with greater transcriptional response. These results demonstrate how fluidic stimuli reveal the cellular phenotype previously masked by severe necrosis. Our platform advances 3D cellular models and enables study into hypoxia modulation, cancer metabolism, and drug screening within pathophysiological conditions.


Subject(s)
Prostatic Neoplasms , Spheroids, Cellular , Humans , Male , Cell Culture Techniques/methods , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Drug Evaluation, Preclinical
5.
J Vis Exp ; (153)2019 11 22.
Article in English | MEDLINE | ID: mdl-31814611

ABSTRACT

The prostate epithelium is comprised predominantly of basal and luminal cells. In vivo lineage tracing has been utilized to define the differentiation capacity of mouse prostate basal and luminal cells during development, tissue-regeneration and transformation. However, evaluating cell-intrinsic and extrinsic regulators of prostate epithelial differentiation capacity using a lineage tracing approach often requires extensive breeding and can be cost-prohibitive. In the prostate organoid assay, basal and luminal cells generate prostatic epithelium ex vivo. Importantly, primary epithelial cells can be isolated from mice of any genetic background or mice treated with any number of small molecules prior to, or after, plating into three-dimensional (3D) culture. Sufficient material for evaluation of differentiation capacity is generated after 7-10 days. Collection of basal-derived and luminal-derived organoids for (1) protein analysis by Western blot and (2) immunohistochemical analysis of intact organoids by whole-mount confocal microscopy enables researchers to evaluate the ex vivo differentiation capacity of prostate epithelial cells. When used in combination, these two approaches provide complementary information about the differentiation capacity of prostate basal and luminal cells in response to genetic or pharmacological manipulation.


Subject(s)
Cell Differentiation , Epithelial Cells/cytology , Prostate/cytology , Animals , Cell Count , Humans , Male , Mice , Organoids/cytology
6.
Cell Rep ; 28(6): 1499-1510.e6, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31390564

ABSTRACT

Aging is associated with loss of tissue mass and a decline in adult stem cell function in many tissues. In contrast, aging in the prostate is associated with growth-related diseases including benign prostatic hyperplasia (BPH). Surprisingly, the effects of aging on prostate epithelial cells have not been established. Here we find that organoid-forming progenitor activity of mouse prostate basal and luminal cells is maintained with age. This is caused by an age-related expansion of progenitor-like luminal cells that share features with human prostate luminal progenitor cells. The increase in luminal progenitor cells may contribute to greater risk for growth-related disease in the aging prostate. Importantly, we demonstrate expansion of human luminal progenitor cells in BPH. In summary, we define a Trop2+ luminal progenitor subset and identify an age-related shift in the luminal compartment of the mouse and human prostate epithelium.


Subject(s)
Aging/pathology , Prostate/pathology , Prostatic Hyperplasia/pathology , Stem Cells/pathology , Adolescent , Adult , Animals , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Cell Proliferation , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Organoids/pathology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...