Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Proc Natl Acad Sci U S A ; 120(42): e2303774120, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37816052

ABSTRACT

Although robustly expressed in the disease-free (DF) breast stroma, CD36 is consistently absent from the stroma surrounding invasive breast cancers (IBCs). In this study, we primarily observed CD36 expression in adipocytes and intralobular capillaries within the DF breast. Larger vessels concentrated in interlobular regions lacked CD36 and were instead marked by the expression of CD31. When evaluated in perilesional capillaries surrounding ductal carcinoma in situ, a nonobligate IBC precursor, CD36 loss was more commonly observed in lesions associated with subsequent IBC. Peroxisome proliferator-activated receptor γ (PPARγ) governs the expression of CD36 and genes involved in differentiation, metabolism, angiogenesis, and inflammation. Coincident with CD36 loss, we observed a dramatic suppression of PPARγ and its target genes in capillary endothelial cells (ECs) and pericytes, which typically surround and support the stability of the capillary endothelium. Factors present in conditioned media from malignant cells repressed PPARγ and its target genes not only in cultured ECs and pericytes but also in adipocytes, which require PPARγ for proper differentiation. In addition, we identified a role for PPARγ in opposing the transition of pericytes toward a tumor-supportive myofibroblast phenotype. In mouse xenograft models, early intervention with rosiglitazone, a PPARγ agonist, demonstrated significant antitumor effects; however, following the development of a palpable tumor, the antitumor effects of rosiglitazone were negated by the repression of PPARγ in the mouse stroma. In summary, PPARγ activity in healthy tissues places several stromal cell types in an antitumorigenic state, directly inhibiting EC proliferation, maintaining adipocyte differentiation, and suppressing the transition of pericytes into tumor-supportive myofibroblasts.


Subject(s)
Breast Neoplasms , Animals , Female , Humans , Mice , Adipocytes/metabolism , Breast Neoplasms/pathology , Endothelial Cells/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Rosiglitazone/pharmacology
2.
Gastroenterology ; 165(2): 374-390, 2023 08.
Article in English | MEDLINE | ID: mdl-37196797

ABSTRACT

BACKGROUND & AIMS: Elements of field cancerization, including atrophic gastritis, metaplasia, and dysplasia, promote gastric cancer development in association with chronic inflammation. However, it remains unclear how stroma changes during carcinogenesis and how the stroma contributes to progression of gastric preneoplasia. Here we investigated heterogeneity of fibroblasts, one of the most important elements in the stroma, and their roles in neoplastic transformation of metaplasia. METHODS: We used single-cell transcriptomics to evaluate the cellular heterogeneity of mucosal cells from patients with gastric cancer. Tissue sections from the same cohort and tissue microarrays were used to identify the geographical distribution of distinct fibroblast subsets. We further evaluated the role of fibroblasts from pathologic mucosa in dysplastic progression of metaplastic cells using patient-derived metaplastic gastroids and fibroblasts. RESULTS: We identified 4 subsets of fibroblasts within stromal cells defined by the differential expression of PDGFRA, FBLN2, ACTA2, or PDGFRB. Each subset was distributed distinctively throughout stomach tissues with different proportions at each pathologic stage. The PDGFRα+ subset expanded in metaplasia and cancer compared with normal, maintaining a close proximity with the epithelial compartment. Co-culture of metaplasia- or cancer-derived fibroblasts with gastroids showing the characteristics of spasmolytic polypeptide-expressing metaplasia-induced disordered growth, loss of metaplastic markers, and increases in markers of dysplasia. Culture of metaplastic gastroids with conditioned media from metaplasia- or cancer-derived fibroblasts also promoted dysplastic transition. CONCLUSIONS: These findings indicate that fibroblast associations with metaplastic epithelial cells can facilitate direct transition of metaplastic spasmolytic polypeptide-expressing metaplasia cell lineages into dysplastic lineages.


Subject(s)
Gastric Mucosa , Stomach Neoplasms , Humans , Gastric Mucosa/pathology , Stomach Neoplasms/pathology , Hyperplasia , Metaplasia/pathology , Fibroblasts/metabolism
3.
Genes Dev ; 30(9): 1002-19, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27151975

ABSTRACT

The tumor stroma is no longer seen solely as physical support for mutated epithelial cells but as an important modulator and even a driver of tumorigenicity. Within the tumor stromal milieu, heterogeneous populations of fibroblast-like cells, collectively termed carcinoma-associated fibroblasts (CAFs), are key players in the multicellular, stromal-dependent alterations that contribute to malignant initiation and progression. This review focuses on novel insights into the contributions of CAFs to disease progression, emergent events leading to the generation of CAFs, identification of CAF-specific biomarkers predictive of disease outcome, and recent therapeutic approaches aimed at blunting or reverting detrimental protumorigenic phenotypes associated with CAFs.


Subject(s)
Carcinoma/pathology , Fibroblasts/pathology , Animals , Carcinoma/physiopathology , Carcinoma/therapy , Disease Progression , Humans
4.
Proteomics ; 23(7-8): e2200021, 2023 04.
Article in English | MEDLINE | ID: mdl-36228107

ABSTRACT

Early events associated with chronic inflammation and cancer involve significant remodeling of the extracellular matrix (ECM), which greatly affects its composition and functional properties. Using lung squamous cell carcinoma (LSCC), a chronic inflammation-associated cancer (CIAC), we optimized a robust proteomic pipeline to discover potential biomarker signatures and protein changes specifically in the stroma. We combined ECM enrichment from fresh human tissues, data-independent acquisition (DIA) strategies, and stringent statistical processing to analyze "Tumor" and matched adjacent histologically normal ("Matched Normal") tissues from patients with LSCC. Overall, 1802 protein groups were quantified with at least two unique peptides, and 56% of those proteins were annotated as "extracellular." Confirming dramatic ECM remodeling during CIAC progression, 529 proteins were significantly altered in the "Tumor" compared to "Matched Normal" tissues. The signature was typified by a coordinated loss of basement membrane proteins and small leucine-rich proteins. The dramatic increase in the stromal levels of SERPINH1/heat shock protein 47, that was discovered using our ECM proteomic pipeline, was validated by immunohistochemistry (IHC) of "Tumor" and "Matched Normal" tissues, obtained from an independent cohort of LSCC patients. This integrated workflow provided novel insights into ECM remodeling during CIAC progression, and identified potential biomarker signatures and future therapeutic targets.


Subject(s)
Carcinoma, Squamous Cell , Proteomics , Humans , Extracellular Matrix/metabolism , Lung/metabolism , Carcinoma, Squamous Cell/pathology , Inflammation/metabolism , Extracellular Matrix Proteins/metabolism
5.
Nature ; 518(7539): 317-30, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25693563

ABSTRACT

The reference human genome sequence set the stage for studies of genetic variation and its association with human disease, but epigenomic studies lack a similar reference. To address this need, the NIH Roadmap Epigenomics Consortium generated the largest collection so far of human epigenomes for primary cells and tissues. Here we describe the integrative analysis of 111 reference human epigenomes generated as part of the programme, profiled for histone modification patterns, DNA accessibility, DNA methylation and RNA expression. We establish global maps of regulatory elements, define regulatory modules of coordinated activity, and their likely activators and repressors. We show that disease- and trait-associated genetic variants are enriched in tissue-specific epigenomic marks, revealing biologically relevant cell types for diverse human traits, and providing a resource for interpreting the molecular basis of human disease. Our results demonstrate the central role of epigenomic information for understanding gene regulation, cellular differentiation and human disease.


Subject(s)
Epigenesis, Genetic/genetics , Epigenomics , Genome, Human/genetics , Base Sequence , Cell Lineage/genetics , Cells, Cultured , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Chromosomes, Human/chemistry , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA Methylation , Datasets as Topic , Enhancer Elements, Genetic/genetics , Genetic Variation/genetics , Genome-Wide Association Study , Histones/metabolism , Humans , Organ Specificity/genetics , RNA/genetics , Reference Values
6.
Mol Cell ; 50(4): 552-64, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23706820

ABSTRACT

Cellular stress results in profound changes in RNA and protein synthesis. How cells integrate this intrinsic, p53-centered program with extracellular signals is largely unknown. We demonstrate that TGF-ß1 signaling interferes with the stress response through coordinate transcriptional and translational repression of p53 levels, which reduces p53-activated transcription, and apoptosis in precancerous cells. Mechanistically, E2F-4 binds constitutively to the TP53 gene and induces transcription. TGF-ß1-activated Smads are recruited to a composite Smad/E2F-4 element by an E2F-4/p107 complex that switches to a Smad corepressor, which represses TP53 transcription. TGF-ß1 also causes dissociation of ribosomal protein RPL26 and elongation factor eEF1A from p53 mRNA, thereby reducing p53 mRNA association with polyribosomes and p53 translation. TGF-ß1 signaling is dominant over stress-induced transcription and translation of p53 and prevents stress-imposed downregulation of Smad proteins. Thus, crosstalk between the TGF-ß and p53 pathways defines a major node of regulation in the cellular stress response, enhancing drug resistance.


Subject(s)
Gene Expression Regulation/drug effects , Stress, Physiological/drug effects , Transforming Growth Factor beta1/pharmacology , Tumor Suppressor Protein p53/genetics , Apoptosis/drug effects , Apoptosis/genetics , Base Sequence , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , E2F4 Transcription Factor/genetics , E2F4 Transcription Factor/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Immunohistochemistry , Mammary Glands, Human/cytology , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Smad Proteins/genetics , Smad Proteins/metabolism , Stress, Physiological/genetics , Tumor Suppressor Protein p53/metabolism
7.
Nature ; 486(7403): 395-9, 2012 Apr 04.
Article in English | MEDLINE | ID: mdl-22495314

ABSTRACT

Primary triple-negative breast cancers (TNBCs), a tumour type defined by lack of oestrogen receptor, progesterone receptor and ERBB2 gene amplification, represent approximately 16% of all breast cancers. Here we show in 104 TNBC cases that at the time of diagnosis these cancers exhibit a wide and continuous spectrum of genomic evolution, with some having only a handful of coding somatic aberrations in a few pathways, whereas others contain hundreds of coding somatic mutations. High-throughput RNA sequencing (RNA-seq) revealed that only approximately 36% of mutations are expressed. Using deep re-sequencing measurements of allelic abundance for 2,414 somatic mutations, we determine for the first time-to our knowledge-in an epithelial tumour subtype, the relative abundance of clonal frequencies among cases representative of the population. We show that TNBCs vary widely in their clonal frequencies at the time of diagnosis, with the basal subtype of TNBC showing more variation than non-basal TNBC. Although p53 (also known as TP53), PIK3CA and PTEN somatic mutations seem to be clonally dominant compared to other genes, in some tumours their clonal frequencies are incompatible with founder status. Mutations in cytoskeletal, cell shape and motility proteins occurred at lower clonal frequencies, suggesting that they occurred later during tumour progression. Taken together, our results show that understanding the biology and therapeutic responses of patients with TNBC will require the determination of individual tumour clonal genotypes.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Evolution, Molecular , Mutation/genetics , Alleles , Breast Neoplasms/diagnosis , Clone Cells/metabolism , Clone Cells/pathology , DNA Copy Number Variations/genetics , DNA Mutational Analysis , Disease Progression , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Genotype , High-Throughput Nucleotide Sequencing , Humans , INDEL Mutation/genetics , Point Mutation/genetics , Precision Medicine , Reproducibility of Results , Sequence Analysis, RNA
8.
Genome Res ; 23(9): 1522-40, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23804400

ABSTRACT

DNA methylation plays key roles in diverse biological processes such as X chromosome inactivation, transposable element repression, genomic imprinting, and tissue-specific gene expression. Sequencing-based DNA methylation profiling provides an unprecedented opportunity to map and compare complete DNA methylomes. This includes one of the most widely applied technologies for measuring DNA methylation: methylated DNA immunoprecipitation followed by sequencing (MeDIP-seq), coupled with a complementary method, methylation-sensitive restriction enzyme sequencing (MRE-seq). A computational approach that integrates data from these two different but complementary assays and predicts methylation differences between samples has been unavailable. Here, we present a novel integrative statistical framework M&M (for integration of MeDIP-seq and MRE-seq) that dynamically scales, normalizes, and combines MeDIP-seq and MRE-seq data to detect differentially methylated regions. Using sample-matched whole-genome bisulfite sequencing (WGBS) as a gold standard, we demonstrate superior accuracy and reproducibility of M&M compared to existing analytical methods for MeDIP-seq data alone. M&M leverages the complementary nature of MeDIP-seq and MRE-seq data to allow rapid comparative analysis between whole methylomes at a fraction of the cost of WGBS. Comprehensive analysis of nineteen human DNA methylomes with M&M reveals distinct DNA methylation patterns among different tissue types, cell types, and individuals, potentially underscoring divergent epigenetic regulation at different scales of phenotypic diversity. We find that differential DNA methylation at enhancer elements, with concurrent changes in histone modifications and transcription factor binding, is common at the cell, tissue, and individual levels, whereas promoter methylation is more prominent in reinforcing fundamental tissue identities.


Subject(s)
Algorithms , DNA Methylation , Genome, Human , Sequence Analysis, DNA/methods , Data Interpretation, Statistical , High-Throughput Nucleotide Sequencing/methods , Humans , Organ Specificity
9.
Proc Natl Acad Sci U S A ; 110(12): 4598-603, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23487770

ABSTRACT

We identified cell surface markers associated with repression of p16(INK4a)/cyclin-dependent kinase inhibitor 2A(CDKN2A), a critical determinant in the acquisition of a plastic state. These cell surface markers allowed direct isolation of rare cells from healthy human breast tissue that exhibit extensive lineage plasticity. This subpopulation is poised to transcribe plasticity markers, OCT3/4, SOX2, and NANOG, at levels similar to those measured in human embryonic stem cells and to acquire a plastic state sensitive to environmental programming. In vitro, in vivo, and teratoma assays demonstrated that either a directly sorted (uncultured) or a single-cell (clonogenic) cell population from primary tissue can differentiate into functional derivatives of each germ layer, ectodermal, endodermal, and mesodermal. In contrast to other cells that express OCT3/4, SOX2, and NANOG, these human endogenous plastic somatic cells are mortal, express low telomerase activity, expand for an extensive but finite number of population doublings, and maintain a diploid karyotype before arresting in G1.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Antigens, Differentiation/biosynthesis , Breast/cytology , Breast/metabolism , Cell Separation/methods , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Female , G1 Phase Cell Cycle Checkpoints/physiology , Homeodomain Proteins/biosynthesis , Humans , Nanog Homeobox Protein , Octamer Transcription Factor-3/biosynthesis , SOXB1 Transcription Factors/biosynthesis
10.
Org Biomol Chem ; 13(45): 11078-86, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26400105

ABSTRACT

Cyclooxygenases (COXs) are enzymes that play a vital role in the inflammatory cascade through the generation of prostaglandins. Their over-expression has been implicated in numerous diseases. In particular, over-expression of COX-2 has been shown to be a predictive biomarker for progression of pre-malignant lesions towards invasive cancer in various tissues. This makes the early detection of COX-2 expressing lesions of high clinical relevance. Herein we describe the development of the first self-immolating trigger which targets COXs. We incorporated our trigger design into 2 activatable fluorogenic probes and demonstrated COX-specific activation in vitro. Experimental data revealed probe activation was likely caused by solvent-exposed amino acids on the surface of the COXs. Overall, the probes reported here mark the first step towards developing self-immolating imaging/therapeutic agents targeted to specific COXs.


Subject(s)
Aspirin/analogs & derivatives , Aspirin/metabolism , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Animals , Cell Line , Cyclooxygenase 1/analysis , Cyclooxygenase 2/analysis , Humans , Mice , Models, Molecular , Optical Imaging , Sheep , Spectrometry, Fluorescence , Swine
11.
Biochem Biophys Res Commun ; 446(2): 434-40, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24607279

ABSTRACT

Membrane skeletal protein 4.1R(80) plays a key role in regulation of erythrocyte plasticity. Protein 4.1R(80) interactions with transmembrane proteins, such as glycophorin C (GPC), are regulated by Ca(2+)-saturated calmodulin (Ca(2+)/CaM) through simultaneous binding to a short peptide (pep11; A(264)KKLWKVCVEHHTFFRL) and a serine residue (Ser(185)), both located in the N-terminal 30 kDa FERM domain of 4.1R(80) (H·R30). We have previously demonstrated that CaM binding to H·R30 is Ca(2+)-independent and that CaM binding to H·R30 is responsible for the maintenance of H·R30 ß-sheet structure. However, the mechanisms responsible for the regulation of CaM binding to H·R30 are still unknown. To investigate this, we took advantage of similarities and differences in the structure of Coracle, the Drosophila sp. homologue of human 4.1R(80), i.e. conservation of the pep11 sequence but substitution of the Ser(185) residue with an alanine residue. We show that the H·R30 homologue domain of Coracle, Cor30, also binds to CaM in a Ca(2+)-independent manner and that the Ca(2+)/CaM complex does not affect Cor30 binding to the transmembrane protein GPC. We also document that both H·R30 and Cor30 bind to phosphatidylinositol-4,5 bisphosphate (PIP2) and other phospholipid species and that that PIP2 inhibits Ca(2+)-free CaM but not Ca(2+)-saturated CaM binding to Cor30. We conclude that PIP2 may play an important role as a modulator of apo-CaM binding to 4.1R(80) throughout evolution.


Subject(s)
Calcium/chemistry , Calmodulin/metabolism , Cytoskeletal Proteins/metabolism , Drosophila/metabolism , Membrane Proteins/metabolism , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Animals , Binding Sites , Calmodulin/chemistry , Cytoskeletal Proteins/chemistry , Humans , Membrane Proteins/chemistry , Phosphatidylinositol 4,5-Diphosphate/chemistry , Protein Binding/drug effects
12.
Cell Mol Gastroenterol Hepatol ; 17(4): 639-656, 2024.
Article in English | MEDLINE | ID: mdl-38199279

ABSTRACT

BACKGROUND & AIMS: Chronic inflammatory illnesses are debilitating and recurrent conditions associated with significant comorbidities, including an increased risk of developing cancer. Extensive tissue remodeling is a hallmark of such illnesses, and is both a consequence and a mediator of disease progression. Despite previous characterization of epithelial and stromal remodeling during inflammatory bowel disease, a complete understanding of its impact on disease progression is lacking. METHODS: A comprehensive proteomic pipeline using data-independent acquisition was applied to decellularized colon samples from the Muc2 knockout (Muc2KO) mouse model of colitis for an in-depth characterization of extracellular matrix remodeling. Unique proteomic profiles of the matrisomal landscape were extracted from prepathologic and overt colitis. Integration of proteomics and transcriptomics data sets extracted from the same murine model produced network maps describing the orchestrating role of matrisomal proteins in tissue remodeling during the progression of colitis. RESULTS: The in-depth proteomic workflow used here allowed the addition of 34 proteins to the known colon matrisomal signature. Protein signatures of prepathologic and pathologic colitic states were extracted, differentiating the 2 states by expression of small leucine-rich proteoglycans. We outlined the role of this class and other matrisomal proteins in tissue remodeling during colitis, as well as the potential for coordinated regulation of cell types by matrisomal ligands. CONCLUSIONS: Our work highlights a central role for matrisomal proteins in tissue remodeling during colitis and defines orchestrating nodes that can be exploited in the selection of therapeutic targets.


Subject(s)
Colitis , Proteomics , Mice , Animals , Extracellular Matrix/metabolism , Colitis/pathology , Chronic Disease , Disease Progression
13.
Exp Cell Res ; 318(13): 1467-79, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22429617

ABSTRACT

The 4.1 proteins are cytoskeletal adaptor proteins that are linked to the control of mechanical stability of certain membranes and to the cellular accumulation and cell surface display of diverse transmembrane proteins. One of the four mammalian 4.1 proteins, 4.1R (80 kDa/120 kDa isoforms), has recently been shown to be required for the normal operation of several ion transporters in the heart (Stagg MA et al. Circ Res, 2008; 103: 855-863). The other three (4.1G, 4.1N and 4.1B) are largely uncharacterised in the heart. Here, we use specific antibodies to characterise their expression, distribution and novel activities in the left ventricle. We detected 4.1R, 4.1G and 4.1N by immunofluorescence and immunoblotting, but not 4.1B. Only one splice variant of 4.1N and 4.1G was seen whereas there are several forms of 4.1R. 4.1N, like 4.1R, was present in intercalated discs, but unlike 4.1R, it was not localised at the lateral plasma membrane. Both 4.1R and 4.1N were in internal structures that, at the level of resolution of the light microscope, were close to the Z-disc (possibly T-tubules). 4.1G was also in intracellular structures, some of which were coincident with sarcoplasmic reticulum. 4.1G existed in an immunoprecipitable complex with spectrin and SERCA2. 80 kDa 4.1R was present in subcellular fractions enriched in intercalated discs, in a complex resistant to solubilization under non-denaturing conditions. At the intercalated disc 4.1R does not colocalise with the adherens junction protein, ß-catenin, but does overlap with the other plasma membrane signalling proteins, the Na/K-ATPase and the Na/Ca exchanger NCX1. We conclude that isoforms of 4.1 proteins are differentially compartmentalised in the heart, and that they form specific complexes with proteins central to cardiomyocyte Ca(2+) metabolism.


Subject(s)
Calcium/metabolism , Microfilament Proteins/metabolism , Myocytes, Cardiac/metabolism , Animals , Cell Compartmentation , Cell Membrane/metabolism , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Heart Ventricles/cytology , Heart Ventricles/metabolism , Homeostasis , Immunoblotting , Intracellular Membranes/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/chemistry , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Microscopy, Fluorescence , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Neuropeptides/chemistry , Neuropeptides/metabolism , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Spectrin/chemistry , Spectrin/metabolism
14.
Biochem J ; 446(3): 427-35, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22731252

ABSTRACT

NHE1 (Na(+)/H(+) exchanger isoform 1) has been reported to be hyperactive in 4.1R-null erythrocytes [Rivera, De Franceschi, Peters, Gascard, Mohandas and Brugnara (2006) Am. J. Physiol. Cell Physiol. 291, C880-C886], supporting a functional interaction between NHE1 and 4.1R. In the present paper we demonstrate that 4.1R binds directly to the NHE1cd (cytoplasmic domain of NHE1) through the interaction of an EED motif in the 4.1R FERM (4.1/ezrin/radixin/moesin) domain with two clusters of basic amino acids in the NHE1cd, K(519)R and R(556)FNKKYVKK, previously shown to mediate PIP(2) (phosphatidylinositol 4,5-bisphosphate) binding [Aharonovitz, Zaun, Balla, York, Orlowski and Grinstein (2000) J. Cell. Biol. 150, 213-224]. The affinity of this interaction (K(d) = 100-200 nM) is reduced in hypertonic and acidic conditions, demonstrating that this interaction is of an electrostatic nature. The binding affinity is also reduced upon binding of Ca(2+)/CaM (Ca(2+)-saturated calmodulin) to the 4.1R FERM domain. We propose that 4.1R regulates NHE1 activity through a direct protein-protein interaction that can be modulated by intracellular pH and Na(+) and Ca(2+) concentrations.


Subject(s)
Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Membrane Proteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Amino Acid Sequence , Animals , Binding Sites , Calcium/metabolism , Calmodulin/metabolism , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Hydrogen-Ion Concentration , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Rats , Sequence Alignment , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/chemistry , Sodium-Hydrogen Exchangers/genetics
15.
bioRxiv ; 2023 Jun 11.
Article in English | MEDLINE | ID: mdl-37333362

ABSTRACT

Esophageal adenocarcinoma arises from Barrett's esophagus, a precancerous metaplastic replacement of squamous by columnar epithelium in response to chronic inflammation. Multi-omics profiling, integrating single-cell transcriptomics, extracellular matrix proteomics, tissue-mechanics and spatial proteomics of 64 samples from 12 patients' paths of progression from squamous epithelium through metaplasia, dysplasia to adenocarcinoma, revealed shared and patient-specific progression characteristics. The classic metaplastic replacement of epithelial cells was paralleled by metaplastic changes in stromal cells, ECM and tissue stiffness. Strikingly, this change in tissue state at metaplasia was already accompanied by appearance of fibroblasts with characteristics of carcinoma-associated fibroblasts and of an NK cell-associated immunosuppressive microenvironment. Thus, Barrett's esophagus progresses as a coordinated multi-component system, supporting treatment paradigms that go beyond targeting cancerous cells to incorporating stromal reprogramming.

16.
Cell Syst ; 13(8): 644-664.e8, 2022 08 17.
Article in English | MEDLINE | ID: mdl-35863345

ABSTRACT

The rise and fall of estrogen and progesterone across menstrual cycles and during pregnancy regulates breast development and modifies cancer risk. How these hormones impact each cell type in the breast remains poorly understood because they act indirectly through paracrine networks. Using single-cell analysis of premenopausal breast tissue, we reveal a network of coordinated transcriptional programs representing the tissue-level response to changing hormone levels. Our computational approach, DECIPHER-seq, leverages person-to-person variability in breast composition and cell state to uncover programs that co-vary across individuals. We use differences in cell-type proportions to infer a subset of programs that arise from direct cell-cell interactions regulated by hormones. Further, we demonstrate that prior pregnancy and obesity modify hormone responsiveness through distinct mechanisms: obesity reduces the proportion of hormone-responsive cells, whereas pregnancy dampens the direct response of these cells to hormones. Together, these results provide a comprehensive map of the cycling human breast.


Subject(s)
Breast , Progesterone , Breast/metabolism , Cell Communication , Estrogens/metabolism , Female , Humans , Obesity/metabolism , Pregnancy , Progesterone/metabolism
17.
Biochem J ; 432(2): 407-16, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20812914

ABSTRACT

Membrane skeletal protein 4.1R is the prototypical member of a family of four highly paralogous proteins that include 4.1G, 4.1N and 4.1B. Two isoforms of 4.1R (4.1R135 and 4.1R80), as well as 4.1G, are expressed in erythroblasts during terminal differentiation, but only 4.1R80 is present in mature erythrocytes. Although the function of 4.1R isoforms in erythroid cells has been well characterized, there is little or no information on the function of 4.1G in these cells. In the present study, we performed detailed characterization of the interaction of 4.1G with various erythroid membrane proteins and the regulation of these interactions by calcium-saturated calmodulin. Like both isoforms of 4.1R, 4.1G bound to band 3, glycophorin C, CD44, p55 and calmodulin. While both 4.1G and 4.1R135 interact with similar affinity with CD44 and p55, there are significant differences in the affinity of their interaction with band 3 and glycophorin C. This difference in affinity is related to the non-conserved N-terminal headpiece region of the two proteins that is upstream of the 30 kDa membrane-binding domain that harbours the binding sites for the various membrane proteins. The headpiece region of 4.1G also contains a high-affinity calcium-dependent calmodulin-binding site that plays a key role in modulating its interaction with various membrane proteins. We suggest that expression of the two paralogues of protein 4.1 with different affinities for band 3 and glycophorin C is likely to play a role in assembly of these two membrane proteins during terminal erythroid differentiation.


Subject(s)
Glutathione Transferase/genetics , Animals , Base Sequence , Calmodulin/metabolism , Cloning, Molecular , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , DNA Primers , Glutathione Transferase/metabolism , Kinetics , Mice , Microfilament Proteins , Models, Molecular , Molecular Sequence Data , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
18.
Cancer Res ; 67(18): 8662-70, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875706

ABSTRACT

Transforming growth factor beta1 (TGFbeta) is a tumor suppressor during the initial stage of tumorigenesis, but it can switch to a tumor promoter during neoplastic progression. Ionizing radiation (IR), both a carcinogen and a therapeutic agent, induces TGFbeta activation in vivo. We now show that IR sensitizes human mammary epithelial cells (HMEC) to undergo TGFbeta-mediated epithelial to mesenchymal transition (EMT). Nonmalignant HMEC (MCF10A, HMT3522 S1, and 184v) were irradiated with 2 Gy shortly after attachment in monolayer culture or treated with a low concentration of TGFbeta (0.4 ng/mL) or double treated. All double-treated (IR + TGFbeta) HMEC underwent a morphologic shift from cuboidal to spindle shaped. This phenotype was accompanied by a decreased expression of epithelial markers E-cadherin, beta-catenin, and ZO-1, remodeling of the actin cytoskeleton, and increased expression of mesenchymal markers N-cadherin, fibronectin, and vimentin. Furthermore, double treatment increased cell motility, promoted invasion, and disrupted acinar morphogenesis of cells subsequently plated in Matrigel. Neither radiation nor TGFbeta alone elicited EMT, although IR increased chronic TGFbeta signaling and activity. Gene expression profiling revealed that double-treated cells exhibit a specific 10-gene signature associated with Erk/mitogen-activated protein kinase (MAPK) signaling. We hypothesized that IR-induced MAPK activation primes nonmalignant HMEC to undergo TGFbeta-mediated EMT. Consistent with this, Erk phosphorylation was transiently induced by irradiation and persisted in irradiated cells treated with TGFbeta, and treatment with U0126, a MAP/Erk kinase (MEK) inhibitor, blocked the EMT phenotype. Together, these data show that the interactions between radiation-induced signaling pathways elicit heritable phenotypes that could contribute to neoplastic progression.


Subject(s)
Breast/drug effects , Breast/radiation effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/radiation effects , Transforming Growth Factor beta/pharmacology , Breast/metabolism , Breast/pathology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Enzyme Activation , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial Cells/radiation effects , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/radiation effects , Mesoderm/drug effects , Mesoderm/pathology , Mesoderm/radiation effects , Mitogen-Activated Protein Kinase Kinases/metabolism
19.
Front Biosci ; 11: 1646-66, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16368544

ABSTRACT

Members of the protein 4.1 family of adapter proteins are expressed in a broad panel of tissues including various epithelia where they likely play an important role in maintenance of cell architecture and polarity and in control of cell proliferation. We have recently characterized the structure and distribution of three members of the protein 4.1 family, 4.1B, 4.1R and 4.1N, in mouse kidney. We describe here binding partners for renal 4.1 proteins, identified through the screening of a rat kidney yeast two-hybrid system cDNA library. The identification of putative protein 4.1-based complexes enables us to envision potential functions for 4.1 proteins in kidney: organization of signaling complexes, response to osmotic stress, protein trafficking, and control of cell proliferation. We discuss the relevance of these protein 4.1-based interactions in kidney physio-pathology in the context of their previously identified functions in other cells and tissues. Specifically, we will focus on renal 4.1 protein interactions with beta amyloid precursor protein (beta-APP), 14-3-3 proteins, and the cell swelling-activated chloride channel pICln. We also discuss the functional relevance of another member of the protein 4.1 superfamily, ezrin, in kidney physio-pathology.


Subject(s)
Blood Proteins/physiology , Epithelium/metabolism , Kidney/metabolism , Microtubule-Associated Proteins/physiology , 14-3-3 Proteins/metabolism , Amyloid beta-Peptides/metabolism , Animals , Ankyrins/chemistry , Cell Proliferation , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , DNA, Complementary/metabolism , Disease Progression , Gene Library , Humans , Kidney/embryology , Membrane Proteins , Mice , Multigene Family , Nephrons/metabolism , Osmosis , Protein Binding , Protein Conformation , Rats , Signal Transduction , Two-Hybrid System Techniques
20.
Cell Signal ; 28(12): 1923-1932, 2016 12.
Article in English | MEDLINE | ID: mdl-27705752

ABSTRACT

Endogenous Plastic Somatic (ePS) cells isolated from adult human tissues exhibit extensive lineage plasticity in vitro and in vivo. Here we visualize these rare ePS cells in a latent state, i.e. lacking SOX2, OCT3/4 and NANOG (SON) expression, in non-diseased breast specimens through immunohistochemical analysis of previously identified ePS-specific biomarkers (CD73+, EpCAM+ and CD90-). We also report a novel mechanism by which these latent ePS cells acquire SON expression and plasticity in vitro. Four extracellular factors are necessary for the acquisition of SON expression and lineage plasticity in ePS cells: adenosine (which is produced by the 5' ecto-nucleotidase CD73 and activates in turn the PKA-dependent IL6/STAT3 pathway through the adenosine receptor ADORA2b), IL6, FGF2 and ACTIVIN A. Blocking any pathway component renders ePS cells incapable of SON expression and lineage plasticity. Notably, hESCs do not use adenosine or IL6 nor they express CD73 or ADORA2b and inhibition of adenosine signaling does not ablate their plasticity. Therefore, the data presented here delineate novel circuitry and physiological signals for accessing SON expression in rare, undifferentiated human cells.


Subject(s)
5'-Nucleotidase/metabolism , Cell Plasticity , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/metabolism , Adenosine/metabolism , Adult , Animals , Autocrine Communication , Biomarkers/metabolism , Breast/metabolism , Cell Differentiation , Cyclic AMP-Dependent Protein Kinases/metabolism , Feeder Cells/cytology , Female , Fibroblast Growth Factor 2/metabolism , GPI-Linked Proteins/metabolism , Human Embryonic Stem Cells/metabolism , Humans , Immunohistochemistry , Interleukin-6/metabolism , Mice , Receptor, Adenosine A2B/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Thy-1 Antigens/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL