Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Proc Natl Acad Sci U S A ; 116(22): 10927-10936, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31085655

ABSTRACT

Cell lineage specification is a tightly regulated process that is dependent on appropriate expression of lineage and developmental stage-specific transcriptional programs. Here, we show that Chromodomain Helicase DNA-binding protein 4 (CHD4), a major ATPase/helicase subunit of Nucleosome Remodeling and Deacetylase Complexes (NuRD) in lymphocytes, is essential for specification of the early B cell lineage transcriptional program. In the absence of CHD4 in B cell progenitors in vivo, development of these cells is arrested at an early pro-B-like stage that is unresponsive to IL-7 receptor signaling and unable to efficiently complete V(D)J rearrangements at Igh loci. Our studies confirm that chromatin accessibility and transcription of thousands of gene loci are controlled dynamically by CHD4 during early B cell development. Strikingly, CHD4-deficient pro-B cells express transcripts of many non-B cell lineage genes, including genes that are characteristic of other hematopoietic lineages, neuronal cells, and the CNS, lung, pancreas, and other cell types. We conclude that CHD4 inhibits inappropriate transcription in pro-B cells. Together, our data demonstrate the importance of CHD4 in establishing and maintaining an appropriate transcriptome in early B lymphopoiesis via chromatin accessibility.


Subject(s)
B-Lymphocytes/metabolism , Cell Lineage/genetics , DNA Helicases/genetics , Lymphopoiesis/genetics , Transcription, Genetic/genetics , Animals , B-Lymphocytes/cytology , Chromatin Assembly and Disassembly/genetics , Gene Expression Regulation/genetics , Mice , Mice, Transgenic
2.
JCI Insight ; 9(1)2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38193534

ABSTRACT

Metastatic breast cancer (mBC) tissue in bone was systematically profiled to define the composition of the tumor microenvironment. Gene expression identified a high myeloid signature of patients with improved survival outcomes. Bone metastases were profiled by spatial proteomics to examine myeloid populations within the stroma that correlated with macrophage functions. Single-cell spatial analysis uncovered macrophage activation in the stroma of mBC bone lesions. Matched BC patient samples of primary breast tumor and bone metastasis tissues were compared for gene expression in the bone, where bone morphogenetic protein 2 (BMP2) was most significantly upregulated. Immune cell changes from breast to bone demonstrated a loss of lymphoid cells but a consistent population of macrophages. BMP-activated macrophages were increased uniquely in bone. Bone marrow-derived macrophage activation coupled with BMP inhibition increased inflammatory responses. Using experimental mouse models of mBC bone metastasis and trained immunity, we found that BMP inhibition restricts progression of metastases early in the macrophage activation state but not after tumors were established in the bone. This study revealed unique myeloid BMP activation states that are distinctly integrated with bone metastases.


Subject(s)
Bone Morphogenetic Proteins , Bone Neoplasms , Breast Neoplasms , Macrophages , Animals , Female , Humans , Mice , Bone and Bones , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Phenotype , Tumor Microenvironment , Bone Morphogenetic Proteins/metabolism
3.
Cancer Rep (Hoboken) ; 6(2): e1707, 2023 02.
Article in English | MEDLINE | ID: mdl-36054271

ABSTRACT

BACKGROUND: Prostate cancer is a common cancer in men that annually results in more than 33 000 US deaths. Mortality from prostate cancer is largely from metastatic disease, reflecting on the great strides in the last century of treatments in care for the localized disease. Metastatic castrate resistant prostate cancer (mCRPC) will commonly travel to the bone, creating unique bone pathology that requires nuanced treatments in those sites with surgical, radio and chemotherapeutic interventions. The bone morphogenetic protein (BMP) pathway has been historically studied in the capacity to regulate the osteogenic nature of new bone. New mineralized bone generation is a frequent and common observation in mCRPC and referred to as blastic bone lesions. Less common are bone destructive lesions that are termed lytic. METHODS: We queried the cancer genome atlas (TCGA) prostate cancer databases for the expression of the BMP pathway and found that distinct gene expression of the ligands, soluble antagonists, receptors, and intracellular mediators were altered in localized versus metastatic disease. Human prostate cancer cell lines have an innate ability to promote blastic- or lytic-like bone lesions and we hypothesized that inhibiting BMP signaling in these cell lines would result in a distinct change in osteogenesis gene expression with BMP inhibition. RESULTS: We found unique and common changes by comparing these cell lines response and unique BMP pathway alterations. We treated human PCa cell lines with distinct bone pathologic phenotypes with the BMP inhibitor DMH1 and found distinct osteogenesis responses. We analyzed distinct sites of metastatic PCa in the TCGA and found that BMP signaling was selectively altered in commons sites such as lymph node, bone and liver compared to primary tumors. CONCLUSIONS: Overall we conclude that BMPs in metastatic prostate cancer are important signals and functional mediators of diverse processes that have potential for individualized precision oncology in mCRPC.


Subject(s)
Osteogenesis , Prostatic Neoplasms, Castration-Resistant , Male , Humans , Precision Medicine , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/pharmacology , Cell Line
4.
Eur J Immunol ; 41(6): 1787-93, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21469119

ABSTRACT

In the absence of early B-cell factor 1 (EBF1), B-cell development is arrested at an uncommitted progenitor stage that exhibits increased lineage potentials. Previously, we investigated the roles of EBF1 and its DNA-binding partner Runx1 by evaluating B lymphopoiesis in single (EBF1(het) and Runx1(het)) and compound haploinsufficent (Ebf1(+/-) Runx1(+/-), ER(het)) mice. Here, we demonstrate that decreased Ebf1 gene dosage results in the inappropriate expression of NK-cell lineage-specific genes in B-cell progenitors. Moreover, prolonged expression of Ly6a/Sca-1 suggested the maintenance of a relatively undifferentiated phenotype. These effects were exacerbated by reduced expression of Runx1 and occurred despite expression of Pax5. Repression of inappropriately expressed genes was restored in most pre-B and all immature B cells of ER(het) mice. Enforced EBF1 expression repressed promiscuous transcription in pro-B cells of ER(het) mice and in Ebf1(-/-) Pax5(-/-) fetal liver cells. Together, our studies suggest that normal levels of EBF1 are critical for maintaining B-cell identity by directing repression of non-B-cell-specific genes.


Subject(s)
B-Lymphocytes/metabolism , Cell Lineage , Lymphopoiesis , Precursor Cells, B-Lymphoid/metabolism , Trans-Activators/metabolism , Animals , Antigens, Differentiation/metabolism , Antigens, Ly/genetics , Antigens, Ly/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Lineage/genetics , Cell Lineage/immunology , Cells, Cultured , Gene Dosage/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Lymphopoiesis/genetics , Lymphopoiesis/immunology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mice, Mutant Strains , PAX5 Transcription Factor/genetics , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/pathology , Trans-Activators/genetics , Trans-Activators/immunology
5.
Front Endocrinol (Lausanne) ; 12: 769316, 2021.
Article in English | MEDLINE | ID: mdl-34956082

ABSTRACT

From the 33,000 men in the U.S. who die from prostate cancer each year, the majority of these patients exhibit metastatic disease with bone being the most common site of metastasis. Prostate cancer bone metastases are commonly blastic, exhibiting new growth of unhealthy sclerotic bone, which can cause painful skeletal related events. Patient's current care entails androgen deprivation therapy, anti-resorptive agents, radiation, and chemotherapy to help control the spread of the cancer but little intervention is available to treat blastic bone disease. The transforming growth factor beta (TGFß) and bone morphogenetic protein (BMP) pathways are known to regulate bone growth and resorption of destructive lytic bone lesions, yet the role of TGFß/BMP signaling in prostate cancer blastic vs lytic bone lesions are not fully understood. We hypothesized that to target the BMP/TGFß pathway, a useful biomarker of bone lytic or blastic pathology would have superior response. We show distinct BMP vs. TGFß signaling in clinical samples of human prostate cancer bone metastases with either lytic or blastic pathologies. BMPs exhibit distinct effects on bone homeostasis, so to examine the effect of BMP inhibition on healthy bone, we treated mice with the BMP receptor small molecule antagonist DMH1 and saw a modest temporary improvement in bone health, with increased trabecular bone. We next sought to use the BMP inhibitor DMH1 to treat bone metastasis engraftment seeded by a caudal artery injection of the lytic human prostate cell line PC3 in immunodeficient mice. The colonization by PC3 cells to the bone were restricted with DMH1 treatment and bone health was importantly preserved. We next proceeded to test BMP inhibition in an injury model of established bone metastasis via intratibial injection of the MYC-CaP mouse prostate cell line into FVBN syngeneic mice. DMH1 treated mice had a modest decrease in trabecular bone and reduced lymphocytes in circulation without affecting tumor growth. Taken together we show unique responses to BMP inhibition in metastatic prostate cancer in the bone. These studies suggest that profiling bone lesions in metastatic prostate cancer can help identify therapeutic targets that not only treat the metastatic tumor but also address the need to better treat the distinct tumor induced bone disease.


Subject(s)
Adenocarcinoma/secondary , Bone Morphogenetic Proteins/metabolism , Bone Neoplasms/secondary , Prostatic Neoplasms/pathology , Signal Transduction/physiology , Adenocarcinoma/metabolism , Animals , Bone Density/drug effects , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Cell Line, Tumor , Disease Models, Animal , Femur/drug effects , Humans , Male , Mice , Prostatic Neoplasms/metabolism , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Quinolines/pharmacology , Quinolines/therapeutic use , Tibia/drug effects , Transforming Growth Factor beta/metabolism
6.
Front Oncol ; 10: 357, 2020.
Article in English | MEDLINE | ID: mdl-32318332

ABSTRACT

The Bone Morphogenetic Protein (BMP) pathway is a member of the TGFß signaling family and has complex roles in cancer. BMP signaling is rarely mutated and can be frequently overexpressed in many human cancers. The dichotomous role of BMPs as both tumor promoters and suppressors appears to be largely context based in both the cancer cell and the surrounding microenvironment. Myeloid cells including macrophages and neutrophils have been shown to be tumor promoting when stimulated from BMPs. We found that conditional deletion of BMPR1a in myeloid cells (LysMCre) restricts tumor progression in a syngeneic mouse prostate cancer model. Specific changes occurred in myeloid cells both in tumor bearing mice and tumor naïve mice throughout multiple tissues. We profiled myeloid subsets in the bone marrow, spleen and primary tumor and found myeloid BMPR1a loss altered the differentiation and lineage capability of distinct populations by histologic, flow cytometry and high dimensional mass cytometry analysis. We further confirmed the requirement for BMP signaling with pharmacologic inhibition of THP-1 and Raw264.7 activated into M2 macrophages with the BMP inhibitor DMH1. M2 polarized primary bone marrow derived cells from LysMCre BMPR1a knockout mice indicated a distinct requirement for BMP signaling in myeloid cells during M2 activation. These results indicate a unique necessity for BMP signaling in myeloid cells during tumor progression.

7.
J Immunother Cancer ; 7(1): 293, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31703602

ABSTRACT

The most common metastatic lesions of prostate cancer are in bone and can be classified into three distinct pathology subtypes: lytic, blastic, and an indeterminate mixture of both. We investigated a cohort of decalcified formalin-fixed and paraffin-embedded (FFPE) patient specimens from the bone that contained metastatic prostate cancer with lytic or blastic features. These tissue sections were utilized for immunohistochemistry (IHC) staining, isolation of RNA for gene expression, and Digital Spatial Profiling (DSP) of changes in both the tumor and microenvironment. A diverse set of unique immune cell populations and signaling pathways to both lytic and blastic types of prostate cancer metastases were present. In blastic lesions immune cells were enriched for pSTAT3 and components of the JAK-STAT pathway. In lytic-type lesions, immune cells were enriched for pAKT activity and components of the PI3K-AKT pathway. Enrichment for immune checkpoints including PD-L1, B7-H4, OX40L, and IDO-1 were identified in blastic prostate cancer, providing new therapeutic targets for patients with bone metastases. Biopsies could guide selection of patients into appropriate therapeutic interventions based on protein levels and RNA expression of desired targets in metastatic disease. Molecular pathology has been an excellent complement to the diagnosis, treatment, and management of primary tumors and could be successfully extended to patients with metastatic lesions.


Subject(s)
Bone Neoplasms/diagnosis , Bone Neoplasms/secondary , Prostatic Neoplasms/pathology , Tumor Microenvironment , Biomarkers , Computational Biology/methods , Gene Expression Profiling , Humans , Immunohistochemistry , Male , Prostatic Neoplasms/etiology , Prostatic Neoplasms/metabolism , Signal Transduction
8.
Mol Cell Biol ; 38(17)2018 09 01.
Article in English | MEDLINE | ID: mdl-29915154

ABSTRACT

Zinc finger protein 521 (ZFP521), a DNA-binding protein containing 30 Krüppel-like zinc fingers, has been implicated in the differentiation of multiple cell types, including hematopoietic stem and progenitor cells (HSPC) and B lymphocytes. Here, we report a novel role for ZFP521 in regulating the earliest stages of hematopoiesis and lymphoid cell development via a cell-extrinsic mechanism. Mice with inactivated Zfp521 genes (Zfp521-/-) possess reduced frequencies and numbers of hematopoietic stem and progenitor cells, common lymphoid progenitors, and B and T cell precursors. Notably, ZFP521 deficiency changes bone marrow microenvironment cytokine levels and gene expression within resident HSPC, consistent with a skewing of hematopoiesis away from lymphopoiesis. These results advance our understanding of ZFP521's role in normal hematopoiesis, justifying further research to assess its potential as a target for cancer therapies.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Stem Cell Niche/physiology , Transcription Factors/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cytokines/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Lymphopoiesis/genetics , Lymphopoiesis/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myelopoiesis/genetics , Myelopoiesis/physiology , Protein Binding , Stem Cell Niche/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL