Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Nat Biotechnol ; 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075149

ABSTRACT

Mass cytometry uses metal-isotope-tagged antibodies to label targets of interest, which enables simultaneous measurements of ~50 proteins or protein modifications in millions of single cells, but its sensitivity is limited. Here, we present a signal amplification technology, termed Amplification by Cyclic Extension (ACE), implementing thermal-cycling-based DNA in situ concatenation in combination with 3-cyanovinylcarbazole phosphoramidite-based DNA crosslinking to enable signal amplification simultaneously on >30 protein epitopes. We demonstrate the utility of ACE in low-abundance protein quantification with suspension mass cytometry to characterize molecular reprogramming during the epithelial-to-mesenchymal transition as well as the mesenchymal-to-epithelial transition. We show the capability of ACE to quantify the dynamics of signaling network responses in human T lymphocytes. We further present the application of ACE in imaging mass cytometry-based multiparametric tissue imaging to identify tissue compartments and profile spatial aspects related to pathological states in polycystic kidney tissues.

2.
Nature ; 632(8025): 603-613, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38987604

ABSTRACT

A broad range of brain pathologies critically relies on the vasculature, and cerebrovascular disease is a leading cause of death worldwide. However, the cellular and molecular architecture of the human brain vasculature remains incompletely understood1. Here we performed single-cell RNA sequencing analysis of 606,380 freshly isolated endothelial cells, perivascular cells and other tissue-derived cells from 117 samples, from 68 human fetuses and adult patients to construct a molecular atlas of the developing fetal, adult control and diseased human brain vasculature. We identify extensive molecular heterogeneity of the vasculature of healthy fetal and adult human brains and across five vascular-dependent central nervous system (CNS) pathologies, including brain tumours and brain vascular malformations. We identify alteration of arteriovenous differentiation and reactivated fetal as well as conserved dysregulated genes and pathways in the diseased vasculature. Pathological endothelial cells display a loss of CNS-specific properties and reveal an upregulation of MHC class II molecules, indicating atypical features of CNS endothelial cells. Cell-cell interaction analyses predict substantial endothelial-to-perivascular cell ligand-receptor cross-talk, including immune-related and angiogenic pathways, thereby revealing a central role for the endothelium within brain neurovascular unit signalling networks. Our single-cell brain atlas provides insights into the molecular architecture and heterogeneity of the developing, adult/control and diseased human brain vasculature and serves as a powerful reference for future studies.


Subject(s)
Brain Neoplasms , Brain , Central Nervous System Vascular Malformations , Endothelial Cells , Fetus , RNA-Seq , Single-Cell Gene Expression Analysis , Female , Humans , Male , Brain/blood supply , Brain/pathology , Brain/embryology , Brain/metabolism , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Cell Communication , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Cells/cytology , Fetus/blood supply , Fetus/cytology , Fetus/embryology , Central Nervous System Vascular Malformations/pathology , HLA-D Antigens/metabolism , Adult , Health
3.
Bioconjug Chem ; 35(1): 80-91, 2024 01 17.
Article in English | MEDLINE | ID: mdl-38112314

ABSTRACT

Mass cytometry permits the high dimensional analysis of complex biological samples; however, some techniques are not yet integrated into the mass cytometry workflow due to reagent availability. The use of self-labeling protein systems, such as HaloTag, are one such application. Here, we describe the design and implementation of the first mass cytometry ligands for use with HaloTag. "Click"-amenable HaloTag warheads were first conjugated onto poly(l-lysine) or poly(acrylic acid) polymers that were then functionalized with diethylenetriaminepentaacetic acid (DTPA) lutetium metal chelates. Kinetic analysis of the HaloTag labeling rates demonstrated that the structure appended to the 1-chlorohexyl warhead was key to success. A construct with a diethylene glycol spacer appended to a benzamide gave similar rates (kobs ∼ 102 M-1 s-1), regardless of the nature of the polymer. Comparison of the polymer with a small molecule chelate having rapid HaloTag labeling kinetics (kobs ∼ 104 M-1 s-1) suggests the polymers significantly reduced the HaloTag labeling rate. HEK293T cells expressing surface-exposed GFP-HaloTag fusions were labeled with the polymeric constructs and 175Lu content measured by cytometry by time-of-flight (CyTOF). Robust labeling was observed; however, significant nonspecific binding of the constructs to cells was also present. Heavily pegylated polymers demonstrated that nonspecific binding could be reduced to allow cells bearing the HaloTag protein to be distinguished from nonexpressing cells.


Subject(s)
Hydrolases , Polymers , Proteins , Humans , Ligands , Kinetics , HEK293 Cells
5.
Nat Commun ; 14(1): 3150, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37258521

ABSTRACT

How the genetic landscape governs a tumor's response to immunotherapy remains poorly understood. To assess the immune-modulatory capabilities of 573 genes associated with altered cytotoxicity in human cancers, here we perform CRISPR/Cas9 screens directly in mouse lung cancer models. We recover the known immune evasion factors Stat1 and Serpinb9 and identify the cancer testis antigen Adam2 as an immune modulator, whose expression is induced by KrasG12D and further elevated by immunotherapy. Using loss- and gain-of-function experiments, we show that ADAM2 functions as an oncogene by restraining interferon and TNF cytokine signaling causing reduced presentation of tumor-associated antigens. ADAM2 also restricts expression of the immune checkpoint inhibitors PDL1, LAG3, TIGIT and TIM3 in the tumor microenvironment, which might explain why ex vivo expanded and adoptively transferred cytotoxic T-cells show enhanced cytotoxic efficacy in ADAM2 overexpressing tumors. Together, direct in vivo CRISPR/Cas9 screens can uncover genetic alterations that control responses to immunotherapies.


Subject(s)
Antineoplastic Agents , Fertilins , Lung Neoplasms , Serpins , Animals , Humans , Male , Mice , Antigens, Neoplasm , Fertilins/genetics , Immunotherapy , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Membrane Proteins/genetics , Serpins/genetics , T-Lymphocytes, Cytotoxic , Tumor Microenvironment
6.
Adv Healthc Mater ; 12(19): e2202422, 2023 07.
Article in English | MEDLINE | ID: mdl-37086259

ABSTRACT

Patient-derived organoids have emerged as a useful tool to model tumour heterogeneity. Scaling these complex culture models while enabling stratified analysis of different cellular sub-populations, however, remains a challenge. One strategy to enable higher throughput organoid cultures is the scaffold-supported platform for organoid-based tissues (SPOT). SPOT allows the generation of flat, thin, and dimensionally-defined microtissues in both 96- and 384-well plate footprints that are compatible with longitudinal image-based readouts. SPOT is currently manufactured manually, however, limiting scalability. In this study, an automation approach to engineer tumour-mimetic 3D microtissues in SPOT using a liquid handler is optimized and comparable within- and between-sample variation to standard manual manufacturing is shown. Further, a liquid handler-supported cell extraction protocol to support single-cell-based end-point analysis using high-throughput flow cytometry and multiplexed cytometry by time of flight is developed. As a proof-of-value demonstration, 3D complex tissues containing different proportions of tumour and stromal cells are generated to probe the reciprocal impact of co-culture. It is also demonstrated that primary patient-derived organoids can be incorporated into the pipeline to capture patient-level tumour heterogeneity. It is envisioned that this automated 96/384-SPOT workflow will provide opportunities for future applications in high-throughput screening for novel personalized therapeutic targets.


Subject(s)
Neoplasms , Humans , Workflow , Coculture Techniques , Neoplasms/pathology , High-Throughput Screening Assays/methods , Automation , Organoids
7.
Cancer Discov ; 12(12): 2930-2953, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36108220

ABSTRACT

Systematically investigating the scores of genes mutated in cancer and discerning disease drivers from inconsequential bystanders is a prerequisite for precision medicine but remains challenging. Here, we developed a somatic CRISPR/Cas9 mutagenesis screen to study 215 recurrent "long-tail" breast cancer genes, which revealed epigenetic regulation as a major tumor-suppressive mechanism. We report that components of the BAP1 and COMPASS-like complexes, including KMT2C/D, KDM6A, BAP1, and ASXL1/2 ("EpiDrivers"), cooperate with PIK3CAH1047R to transform mouse and human breast epithelial cells. Mechanistically, we find that activation of PIK3CAH1047R and concomitant EpiDriver loss triggered an alveolar-like lineage conversion of basal mammary epithelial cells and accelerated formation of luminal-like tumors, suggesting a basal origin for luminal tumors. EpiDriver mutations are found in ∼39% of human breast cancers, and ∼50% of ductal carcinoma in situ express casein, suggesting that lineage infidelity and alveogenic mimicry may significantly contribute to early steps of breast cancer etiology. SIGNIFICANCE: Infrequently mutated genes comprise most of the mutational burden in breast tumors but are poorly understood. In vivo CRISPR screening identified functional tumor suppressors that converged on epigenetic regulation. Loss of epigenetic regulators accelerated tumorigenesis and revealed lineage infidelity and aberrant expression of alveogenesis genes as potential early events in tumorigenesis. This article is highlighted in the In This Issue feature, p. 2711.


Subject(s)
Breast Neoplasms , Carcinoma, Intraductal, Noninfiltrating , Humans , Mice , Animals , Female , Breast Neoplasms/pathology , Epigenesis, Genetic , Neoplasm Recurrence, Local/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Cell Transformation, Neoplastic/genetics
8.
Cell Rep Med ; 3(4): 100604, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35492239

ABSTRACT

Localized prostate cancer exhibits multiple genomic alterations and heterogeneity at the proteomic level. Single-cell technologies capture important cell-to-cell variability responsible for heterogeneity in biomarker expression that may be overlooked when molecular alterations are based on bulk tissue samples. This study aims to identify prognostic biomarkers and describe the heterogeneity of prostate cancer and the associated microenvironment by simultaneously quantifying 36 proteins using single-cell mass cytometry analysis of over 1.6 million cells from 58 men with localized prostate cancer. We perform this task, using a high-dimensional clustering pipeline named Franken to describe subpopulations of immune, stromal, and prostate cells, including changes occurring in tumor tissues and high-grade disease that provide insights into the coordinated progression of prostate cancer. Our results further indicate that men with localized disease already harbor rare subpopulations that typically occur in castration-resistant and metastatic disease.


Subject(s)
Prostatic Neoplasms , Proteomics , Genomics , Humans , Male , Orchiectomy , Prostate/metabolism , Prostatic Neoplasms/genetics , Proteomics/methods , Tumor Microenvironment/genetics
9.
Cell Syst ; 12(12): 1173-1186.e5, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34536381

ABSTRACT

A major challenge in the analysis of highly multiplexed imaging data is the assignment of cells to a priori known cell types. Existing approaches typically solve this by clustering cells followed by manual annotation. However, these often require several subjective choices and cannot explicitly assign cells to an uncharacterized type. To help address these issues we present Astir, a probabilistic model to assign cells to cell types by integrating prior knowledge of marker proteins. Astir uses deep recognition neural networks for fast inference, allowing for annotations at the million-cell scale in the absence of a previously annotated reference. We apply Astir to over 2.4 million cells from suspension and imaging datasets and demonstrate its scalability, robustness to sample composition, and interpretable uncertainty estimates. We envision deployment of Astir either for a first broad cell type assignment or to accurately annotate cells that may serve as biomarkers in multiple disease contexts. A record of this paper's transparent peer review process is included in the supplemental information.


Subject(s)
Neural Networks, Computer , Proteomics , Cluster Analysis
11.
Nature ; 578(7796): 615-620, 2020 02.
Article in English | MEDLINE | ID: mdl-31959985

ABSTRACT

Single-cell analyses have revealed extensive heterogeneity between and within human tumours1-4, but complex single-cell phenotypes and their spatial context are not at present reflected in the histological stratification that is the foundation of many clinical decisions. Here we use imaging mass cytometry5 to simultaneously quantify 35 biomarkers, resulting in 720 high-dimensional pathology images of tumour tissue from 352 patients with breast cancer, with long-term survival data available for 281 patients. Spatially resolved, single-cell analysis identified the phenotypes of tumour and stromal single cells, their organization and their heterogeneity, and enabled the cellular architecture of breast cancer tissue to be characterized on the basis of cellular composition and tissue organization. Our analysis reveals multicellular features of the tumour microenvironment and novel subgroups of breast cancer that are associated with distinct clinical outcomes. Thus, spatially resolved, single-cell analysis can characterize intratumour phenotypic heterogeneity in a disease-relevant manner, with the potential to inform patient-specific diagnosis.


Subject(s)
Breast Neoplasms/pathology , Molecular Imaging , Single-Cell Analysis , Biomarkers, Tumor/analysis , Breast Neoplasms/classification , Breast Neoplasms/diagnosis , Humans , Kaplan-Meier Estimate , Phenotype , Proportional Hazards Models , Survival Rate , Tumor Microenvironment
12.
Nat Cancer ; 1(2): 163-175, 2020 02.
Article in English | MEDLINE | ID: mdl-35122013

ABSTRACT

Genomic alterations shape cell phenotypes and the structure of tumor ecosystems in poorly defined ways. To investigate these relationships, we used imaging mass cytometry to quantify the expression of 37 proteins with subcellular spatial resolution in 483 tumors from the METABRIC cohort. Single-cell analysis revealed cell phenotypes spanning epithelial, stromal and immune types. Distinct combinations of cell phenotypes and cell-cell interactions were associated with genomic subtypes of breast cancer. Epithelial luminal cell phenotypes separated into those predominantly impacted by mutations and those affected by copy number aberrations. Several features of tumor ecosystems, including cellular neighborhoods, were linked to prognosis, illustrating their clinical relevance. In summary, systematic analysis of single-cell phenotypic and spatial correlates of genomic alterations in cancer revealed how genomes shape both the composition and architecture of breast tumor ecosystems and will enable greater understanding of the phenotypic impact of genomic alterations.


Subject(s)
Breast Neoplasms , Breast Neoplasms/diagnosis , Ecosystem , Female , Genomics/methods , Humans , Image Cytometry , Prognosis
13.
J Clin Invest ; 128(10): 4525-4542, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30222135

ABSTRACT

The E3 ubiquitin ligase RNF8 plays critical roles in maintaining genomic stability by promoting the repair of DNA double-strand breaks (DSBs) through ubiquitin signaling. Abnormal activation of Notch signaling and defective repair of DSBs promote breast cancer risk. Here, we found that low expression of the full-length RNF8 correlated with poor prognosis for breast cancer patients. Our data revealed that in addition to its role in the repair of DSBs, RNF8 regulated Notch1 signaling and cell-fate determination of mammary luminal progenitors. Mechanistically, RNF8 acted as a negative regulator of Notch signaling by ubiquitylating the active NOTCH1 protein (N1ICD), leading to its degradation. Consistent with abnormal activation of Notch signaling and impaired repair of DSBs in Rnf8-mutant mammary epithelial cells, we observed increased risk of mammary tumorigenesis in mouse models for RNF8 deficiency. Notably, deficiency of RNF8 sensitized breast cancer cells to combination of pharmacological inhibitors of Notch signaling and poly(ADP-ribose) polymerase (PARP), suggesting implications for treatment of breast cancer associated with impaired RNF8 expression or function.


Subject(s)
Carcinogenesis/metabolism , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/metabolism , Neoplasm Proteins/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/biosynthesis , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , DNA Breaks, Double-Stranded , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Receptor, Notch1/genetics , Ubiquitin-Protein Ligases/genetics
14.
Nat Methods ; 14(9): 873-876, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28783155

ABSTRACT

Single-cell, spatially resolved omics analysis of tissues is poised to transform biomedical research and clinical practice. We have developed an open-source, computational histology topography cytometry analysis toolbox (histoCAT) to enable interactive, quantitative, and comprehensive exploration of individual cell phenotypes, cell-cell interactions, microenvironments, and morphological structures within intact tissues. We highlight the unique abilities of histoCAT through analysis of highly multiplexed mass cytometry images of human breast cancer tissues.


Subject(s)
Cell Communication/physiology , Flow Cytometry/methods , Molecular Imaging/methods , Proteome/metabolism , Software , Tissue Array Analysis/methods , Algorithms , Image Interpretation, Computer-Assisted/methods , User-Computer Interface
15.
Nat Rev Cancer ; 17(1): 38-53, 2017 01.
Article in English | MEDLINE | ID: mdl-27932800

ABSTRACT

A compelling long-term goal of cancer biology is to understand the crucial players during tumorigenesis in order to develop new interventions. Here, we review how the four non-redundant tissue inhibitors of metalloproteinases (TIMPs) regulate the pericellular proteolysis of a vast range of matrix and cell surface proteins, generating simultaneous effects on tumour architecture and cell signalling. Experimental studies demonstrate the contribution of TIMPs to the majority of cancer hallmarks, and human cancers invariably show TIMP deregulation in the tumour or stroma. Of the four TIMPs, TIMP1 overexpression or TIMP3 silencing is consistently associated with cancer progression or poor patient prognosis. Future efforts will align mouse model systems with changes in TIMPs in patients, will delineate protease-independent TIMP function, will pinpoint therapeutic targets within the TIMP-metalloproteinase-substrate network and will use TIMPs in liquid biopsy samples as biomarkers for cancer prognosis.


Subject(s)
Neoplasms/metabolism , Neoplasms/physiopathology , Tissue Inhibitor of Metalloproteinases/metabolism , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/physiology , Humans , Proteolysis , Signal Transduction , Tissue Inhibitor of Metalloproteinases/physiology , Tumor Microenvironment/physiology
16.
Mol Cell Oncol ; 3(3): e975082, 2016 May.
Article in English | MEDLINE | ID: mdl-27314104

ABSTRACT

The tumor stroma has the capacity to drive cancer progression, although the mechanisms governing these effects are incompletely understood. Recently, we reported that deletion of tissue inhibitor of metalloproteinases (Timps) in fibroblasts unleashes the function of cancer-associated fibroblasts and identifies a novel mode of stromal-tumor communication that activates key oncogenic pathways invoving Notch and ras homolog gene family, member A (RhoA) via stromal exosomes.

17.
Stem Cell Reports ; 5(1): 31-44, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26095608

ABSTRACT

Systemic and local signals must be integrated by mammary stem and progenitor cells to regulate their cyclic growth and turnover in the adult gland. Here, we show RANK-positive luminal progenitors exhibiting WNT pathway activation are selectively expanded in the human breast during the progesterone-high menstrual phase. To investigate underlying mechanisms, we examined mouse models and found that loss of RANK prevents the proliferation of hormone receptor-negative luminal mammary progenitors and basal cells, an accompanying loss of WNT activation, and, hence, a suppression of lobuloalveologenesis. We also show that R-spondin1 is depleted in RANK-null progenitors, and that its exogenous administration rescues key aspects of RANK deficiency by reinstating a WNT response and mammary cell expansion. Our findings point to a novel role of RANK in dictating WNT responsiveness to mediate hormone-induced changes in the growth dynamics of adult mammary cells.


Subject(s)
Mammary Glands, Animal/metabolism , Receptor Activator of Nuclear Factor-kappa B/genetics , Stem Cells/cytology , Thrombospondins/genetics , Animals , Cell Proliferation/genetics , Female , Humans , Mammary Glands, Animal/growth & development , Mice , Receptor Activator of Nuclear Factor-kappa B/antagonists & inhibitors , Thrombospondins/biosynthesis , Wnt Signaling Pathway/genetics
18.
PLoS One ; 10(3): e0120107, 2015.
Article in English | MEDLINE | ID: mdl-25807548

ABSTRACT

Timp3 is commonly silenced in breast cancer, but mechanistic studies have identified both tumor promotion and suppression effects of this gene. We have taken a genetic approach to determine the impact of Timp3 loss on two mouse models of breast cancer. Interestingly, MMTV-PyMT Timp3-/- mice have delayed tumor onset and 36% of MMTV-Neu Timp3-/- mice remain tumor free. TIMP3 is a regulator of TNF signaling and similar to Timp3, Tnf or Tnfr1 loss delays early tumorigenesis. The tumor suppression in Timp3 null mice requires Tnfr1, but does not result in alterations in the local immune compartment. In the mammary gland, Timps are highly expressed in the stroma and through the transplantation of tumor cells we observe that Timp3 deficiency in the host is sufficient to delay the growth of early, but not advanced tumor cells. Together our data is the first to identify a tumor promoting role of endogenous Timp3 in vivo, the spatial and temporal windows of this effect, and its dependence on Tnfr1.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/genetics , Tissue Inhibitor of Metalloproteinase-3/deficiency , Tissue Inhibitor of Metalloproteinase-3/genetics , Animals , Cell Transformation, Neoplastic/pathology , Female , Male , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/genetics
19.
Nat Cell Biol ; 17(3): 217-27, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25706237

ABSTRACT

Age is the primary risk factor for breast cancer in women. Bipotent basal stem cells actively maintain the adult mammary ductal tree, but with age tissues atrophy. We show that cell-extrinsic factors maintain the adult stem cell pool during ageing and dictate tissue stoichiometry. Mammary stem cells spontaneously expand more than 11-fold in virgin adult female mice lacking specific genes for TIMPs, the natural metalloproteinase inhibitors. Compound Timp1/Timp3 null glands exhibit Notch activation and accelerated gestational differentiation. Proteomics of mutant basal cells uncover altered cytoskeletal and extracellular protein repertoires, and we identify aberrant mitotic spindle orientation in these glands, a process that instructs asymmetric cell division and fate. We find that progenitor activity normally declines with age, but enriched stem/progenitor pools prevent tissue regression in Timp mutant mammary glands without affecting carcinogen-induced cancer susceptibility. Thus, improved stem cell content can extend mouse mammary tissue lifespan without altering cancer risk in this mouse model.


Subject(s)
Aging/genetics , Cell Proliferation/genetics , Mammary Glands, Animal/cytology , Stem Cells/cytology , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-3/genetics , Age Factors , Aging/metabolism , Animals , Cell Differentiation , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Female , Gene Expression Regulation, Developmental , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mice , Mice, Knockout , Mitosis , Receptors, Notch/genetics , Receptors, Notch/metabolism , Risk Factors , Signal Transduction , Spindle Apparatus/metabolism , Spindle Apparatus/pathology , Stem Cells/metabolism , Tissue Inhibitor of Metalloproteinase-1/deficiency , Tissue Inhibitor of Metalloproteinase-3/deficiency
20.
Stem Cell Reports ; 4(3): 313-322, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-28447939

ABSTRACT

Progesterone drives mammary stem and progenitor cell dynamics through paracrine mechanisms that are currently not well understood. Here, we demonstrate that CXCR4, the receptor for stromal-derived factor 1 (SDF-1; CXC12), is a crucial instructor of hormone-induced mammary stem and progenitor cell function. Progesterone elicits specific changes in the transcriptome of basal and luminal mammary epithelial populations, where CXCL12 and CXCR4 represent a putative ligand-receptor pair. In situ, CXCL12 localizes to progesterone-receptor-positive luminal cells, whereas CXCR4 is induced in both basal and luminal compartments in a progesterone-dependent manner. Pharmacological inhibition of CXCR4 signaling abrogates progesterone-directed expansion of basal (CD24+CD49fhi) and luminal (CD24+CD49flo) subsets. This is accompanied by a marked reduction in CD49b+SCA-1- luminal progenitors, their functional capacity, and lobuloalveologenesis. These findings uncover CXCL12 and CXCR4 as novel paracrine effectors of hormone signaling in the adult mammary gland, and present a new avenue for potentially targeting progenitor cell growth and malignant transformation in breast cancer.

SELECTION OF CITATIONS
SEARCH DETAIL