Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Pers Med ; 12(9)2022 Sep 16.
Article in English | MEDLINE | ID: mdl-36143304

ABSTRACT

Endometriosis is a common gynaecological condition characterised by the growth of endometrial tissue outside the uterus and is associated with pain and infertility. Currently, the gold standard for endometriosis diagnosis is laparoscopic excision and histological identification of endometrial epithelial and stromal cells. There is, however, currently no known association between the histological appearance, size, morphology, or subtype of endometriosis and disease prognosis. In this study, we used histopathological software to identify and quantify the number of endometrial epithelial and stromal cells within excised endometriotic lesions and assess the relationship between the cell contents and lesion subtypes. Prior to surgery for suspected endometriosis, patients provided menstrual and abdominal pain and dyspareunia scores. Endometriotic lesions removed during laparoscopic surgery were collected and prepared for immunohistochemistry from 26 patients. Endometrial epithelial and stromal cells were identified with Cytokeratin and CD10 antibodies, respectively. Whole slide sections were digitised and the QuPath software was trained to automatically detect and count epithelial and stromal cells across the whole section. Using this classifier, we identified a significantly larger number of strongly labelled CD10 stromal cells (p = 0.0477) in deeply infiltrating lesions (99,970 ± 2962) compared to superficial lesions (2456 ± 859). We found the ratio of epithelial to stromal cells was inverted in deeply infiltrating endometriosis lesions compared to superficial peritoneal and endometrioma lesions and we subsequently identified a correlation between total endometrial cells and abdominal pain (p = 0.0005) when counted via the automated software. Incorporating histological software into current standard diagnostic pipelines may improve endometriosis diagnosis and provide prognostic information in regards to severity and symptoms and eventually provide the potential to personalise adjuvant treatment decisions.

2.
EBioMedicine ; 73: 103664, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34740105

ABSTRACT

BACKGROUND: Although T cell abundance in solid tumours is associated with better outcomes, it also correlates with a stroma-mediated source of immune suppression driven by TGFß1 and poor overall survival. Whether this also is observed in non-small cell lung cancer (NSCLC) is unknown. METHODS: We utilized molecular analysis of The Cancer Genome Atlas (TCGA) NSLCC cohort to correlate immune activation (IA) gene expression and extracellular matrix/stromal (ECM/stromal) gene expression with patient survival. In an independent cohort of NSCLC samples, we used flow cytometry to identify mesenchymal subsets and ex vivo functional studies to characterize their immune regulatory function. FINDINGS: We observed a high enrichment in a core set of genes defining an IA gene expression signature in NSCLC across TCGA Pan-cancer cohort. High IA signature score correlates with enrichment of ECM/stromal gene signature across TCGA NSCLC datasets. Importantly, a higher ratio of ECM/stromal to IA gene signature score was associated with shorter overall survival. In tumours resected from a separate cohort of NSCLC patients, we identified CD90+CD73+ peritumoral cells that were enriched in the ECM/stromal gene signature, which was amplified by TGFß1. IFNγ and TNFα-primed peritumoral CD90+CD73+ cells upregulate immune checkpoint molecules PD-L1 and IDO1 and secrete an array of cytokines/chemokines including TGFß1. Finally, immune primed peritumoral CD90+CD73+ cells suppress T cell function, which was relieved following combined blockade of PD-L1 and TGFß1 with IDO1 inhibition but not PD-L1 or anti-CD73 alone. INTERPRETATION: Our findings suggest that targeting PD-L1 together with independent biological features of the stroma may enhance host antitumor immunity in NSCLC. FUNDING: LW and HY are supported by a 4-year China Scholarship Council award. This work was funded, in part, by a grant from the Cancer League of Bern, Switzerland to SRRH. Laser scanning microscopy imaging was funded by the R'Equip grant from the Swiss National Science Foundation Nr. 316030_145003.


Subject(s)
5'-Nucleotidase/metabolism , Carcinoma, Non-Small-Cell Lung/etiology , Carcinoma, Non-Small-Cell Lung/metabolism , Immunomodulation , Lung Neoplasms/etiology , Lung Neoplasms/metabolism , Thy-1 Antigens/metabolism , Tumor Microenvironment , Biomarkers , Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung/pathology , Computational Biology , Databases, Genetic , Extracellular Matrix , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Immunomodulation/genetics , Immunophenotyping , Lung Neoplasms/pathology , Models, Biological , Stromal Cells/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Transcriptome , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
3.
Cell Rep Med ; 2(12): 100456, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34751258

ABSTRACT

The ongoing SARS-CoV-2 pandemic continues to lead to high morbidity and mortality. During pregnancy, severe maternal and neonatal outcomes and placental pathological changes have been described. We evaluate SARS-CoV-2 infection at the maternal-fetal interface using precision-cut slices (PCSs) of human placenta. Remarkably, exposure of placenta PCSs to SARS-CoV-2 leads to a full replication cycle with infectious virus release. Moreover, the susceptibility of placental tissue to SARS-CoV-2 replication relates to the expression levels of ACE2. Viral proteins and/or viral RNA are detected in syncytiotrophoblasts, cytotrophoblasts, villous stroma, and possibly Hofbauer cells. While SARS-CoV-2 infection of placenta PCSs does not cause a detectable cytotoxicity or a pro-inflammatory cytokine response, an upregulation of one order of magnitude of interferon type III transcripts is measured. In conclusion, our data demonstrate the capacity of SARS-CoV-2 to infect and propagate in human placenta and constitute a basis for further investigation of SARS-CoV-2 biology at the maternal-fetal interface.


Subject(s)
Placenta/virology , SARS-CoV-2/physiology , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/transmission , COVID-19/virology , Chorionic Villi/virology , Female , Humans , Infectious Disease Transmission, Vertical , Interferons/metabolism , Placenta/cytology , Placenta/metabolism , Pregnancy , RNA, Viral/metabolism , Trophoblasts/cytology , Trophoblasts/virology , Viral Proteins/metabolism , Virus Release , Virus Replication , Interferon Lambda
4.
J Steroid Biochem Mol Biol ; 212: 105943, 2021 09.
Article in English | MEDLINE | ID: mdl-34144151

ABSTRACT

Endometriosis is a common, estrogen-dependent disease, in which endometrial tissue grows in the peritoneal cavity. These lesions often express low levels of progesterone receptors (PR), which potentially play an important role in the insufficient response to progestin treatment. Here, we uncover an interconnection between the downregulated PR expression and the epithelial-to-mesenchymal transition (EMT) in endometriotic lesions. The majority of ectopic epithelial glands (93.1 %, n = 67/72) display heterogeneous states of EMT by immunohistochemistry staining. Interestingly, low PR expression associated with high N-cadherin expression, a hallmark of EMT. In order to gain mechanistic insights, we performed in vitro functional assays with the endometriotic epithelial cell lines EM'osis and 12Z. TGF-ß-induced EMT, marked by elevations of CDH2 and SNAI1/2, led to a significant downregulation of PR gene expression in both cell lines. In contrast, silencing of SNAI1 in EM'osis and of SNAI1 plus SNAI2 in 12Z elevated PR gene expression significantly. We found that not only in vitro, but also in the epithelial component of endometriotic lesions strong expression of SNAI1/2 concurred with weak expression of PR. In summary, these results suggested the negative correlation association of the heterogeneous states of EMT and suppressed PR expression in endometriotic lesions. Our functional assays indicate that EMT contributes to the downregulation of PR expression via the upregulation of EMT-TFs, like SNAI1 and SNAI2, which may ultimately lead to progesterone resistance.


Subject(s)
Endometriosis/metabolism , Epithelial-Mesenchymal Transition , Receptors, Progesterone/genetics , Antigens, CD/metabolism , Cadherins/metabolism , Cell Line , Down-Regulation , Epithelial Cells/metabolism , Female , Humans , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism
5.
Am J Physiol Lung Cell Mol Physiol ; 319(5): L794-L809, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32726135

ABSTRACT

Lung injury in mice induces mobilization of discrete subsets of epithelial progenitor cells to promote new airway and alveolar structures. However, whether similar cell types exist in human lung remains unresolved. Using flow cytometry, we identified a distinct cluster of cells expressing the epithelial cell adhesion molecule (EpCAM), a cell surface marker expressed on epithelial progenitor cells, enriched in the ecto-5'-nucleotidase CD73 in unaffected postnatal human lungs resected from pediatric patients with congenital lung lesions. Within the EpCAM+CD73+ population, a small subset coexpresses integrin ß4 and HTII-280. This population remained stable with age. Spatially, EpCAM+CD73+ cells were positioned along the basal membrane of respiratory epithelium and alveolus next to CD73+ cells lacking EpCAM. Expanded EpCAM+CD73+ cells give rise to a pseudostratified epithelium in a two-dimensional air-liquid interface or a clonal three-dimensional organoid assay. Organoids generated under alveolar differentiation conditions were cystic-like and lacked robust alveolar mature cell types. Compared with unaffected postnatal lung, congenital lung lesions were marked by clusters of EpCAM+CD73+ cells in airway and cystic distal lung structures lined by simple epithelium composed of EpCAM+SCGB1A1+ cells and hyperplastic EpCAM+proSPC+ cells. In non-small-cell lung cancer (NSCLC), there was a marked increase in EpCAM+CD73+ tumor cells enriched in inhibitory immune checkpoint molecules CD47 and programmed death-ligand 1 (PD-L1), which was associated with poor survival in lung adenocarcinoma (LUAD). In conclusion, EpCAM+CD73+ cells are rare novel epithelial progenitor cells in the human lung. Importantly, reemergence of CD73 in lung adenocarcinoma enriched in negative immune checkpoint molecules may serve as a novel therapeutic target.


Subject(s)
5'-Nucleotidase/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Epithelial Cells/metabolism , Stem Cells/cytology , Animals , Cell Differentiation/physiology , Epithelial Cell Adhesion Molecule/metabolism , Humans , Lung/pathology , Lung Neoplasms/metabolism , Mesenchymal Stem Cells/metabolism , Mice
6.
Am J Physiol Lung Cell Mol Physiol ; 318(4): L813-L830, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32073879

ABSTRACT

Our understanding of mesenchymal cell subsets and their function in human lung affected by aging and in certain disease settings remains poorly described. We use a combination of flow cytometry, prospective cell-sorting strategies, confocal imaging, and modeling of microvessel formation using advanced microfluidic chip technology to characterize mesenchymal cell subtypes in human postnatal and adult lung. Tissue was obtained from patients undergoing elective surgery for congenital pulmonary airway malformations (CPAM) and other airway abnormalities including chronic obstructive pulmonary disease (COPD). In microscopically normal postnatal human lung, there was a fivefold higher mesenchymal compared with epithelial (EpCAM+) fraction, which diminished with age. The mesenchymal fraction composed of CD90+ and CD90+CD73+ cells was enriched in CXCL12 and platelet-derived growth factor receptor-α (PDGFRα) and located in close proximity to EpCAM+ cells in the alveolar region. Surprisingly, alveolar organoids generated from EpCAM+ cells supported by CD90+ subset were immature and displayed dysplastic features. In congenital lung lesions, cystic air spaces and dysplastic alveolar regions were marked with an underlying thick interstitium composed of CD90+ and CD90+PDGFRα+ cells. In postnatal lung, a subset of CD90+ cells coexpresses the pericyte marker CD146 and supports self-assembly of perfusable microvessels. CD90+CD146+ cells from COPD patients fail to support microvessel formation due to fibrinolysis. Targeting the plasmin-plasminogen system during microvessel self-assembly prevented fibrin gel degradation, but microvessels were narrower and excessive contraction blocked perfusion. These data provide important new information regarding the immunophenotypic identity of key mesenchymal lineages and their change in a diverse setting of congenital lung lesions and COPD.


Subject(s)
Immunomodulation/immunology , Mesenchymal Stem Cells/metabolism , Thy-1 Antigens/immunology , Thy-1 Antigens/metabolism , Adolescent , Biomarkers/metabolism , CD146 Antigen/immunology , CD146 Antigen/metabolism , Cell Separation/methods , Child , Child, Preschool , Epithelial Cell Adhesion Molecule/immunology , Epithelial Cell Adhesion Molecule/metabolism , Female , Humans , Immunologic Factors/immunology , Immunologic Factors/metabolism , Infant , Infant, Newborn , Male , Mesenchymal Stem Cells/immunology , Microvessels/immunology , Microvessels/metabolism , Pericytes/immunology , Pericytes/metabolism , Prospective Studies
7.
Nanotoxicology ; 14(1): 77-96, 2020 02.
Article in English | MEDLINE | ID: mdl-31556347

ABSTRACT

With substantial progress of nanotechnology, there is rising concern about possible adverse health effects related to inhalation of nanomaterials, such as multi-walled carbon nanotubes (MWCNT). In particular, individuals with chronic respiratory disorders, such as chronic obstructive pulmonary disease (COPD), may potentially be more susceptible to adverse health effects related to inhaled MWCNT. Hazard assessment of such inhaled nanomaterials therefore requires timely clarification. This was assessed in this study using a mouse model of COPD by exposing animals to 0.08 µg/cm2 of MWCNT administered by intratracheal instillation. Treatment with MWCNT induced an accumulation of alveolar macrophages (AMφ) in bronchoalveolar lavage fluid (BALF) in COPD mice that increased from 24 h to 7 d. In COPD mice, MWCNT induced a dynamic shift in macrophage polarization as measured by expression of CD38 and CD206, and increased AMφ and lung parenchyma macrophage (LPMΦ) activation with upregulation of co-stimulatory markers CD40 and CD80. Moreover, MWCNT treatment increased the frequencies of pulmonary dendritic cells (DC), leading to an expansion of the CD11b+CD103- DC subset. Although MWCNT did not trigger lung functional or structural changes, they induced an increased expression of the muc5AC transcript in mice with COPD. Our data provide initial evidence that inhaled MWCNT affect the pulmonary mucosal immune system by altering the numbers, phenotype, and activation status of antigen-presenting cell populations. Extrapolating these in vivo mouse findings to human pulmonary MWCNT exposure, caution is warranted in limiting exposure when handling inhalable nanofibers.


Subject(s)
Dendritic Cells/drug effects , Lung/drug effects , Macrophages, Alveolar/drug effects , Nanotubes, Carbon/toxicity , Pulmonary Disease, Chronic Obstructive/chemically induced , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Dendritic Cells/immunology , Disease Models, Animal , Female , Inhalation Exposure , Lung/immunology , Lung/pathology , Macrophages, Alveolar/immunology , Male , Mice, Inbred C57BL , Nanotubes, Carbon/chemistry , Pulmonary Disease, Chronic Obstructive/immunology
8.
Bone ; 97: 278-286, 2017 04.
Article in English | MEDLINE | ID: mdl-28159709

ABSTRACT

Ischemia/reperfusion (I/R) injury has been extensively studied in organs such as heart, brain, liver, kidney, and lung. As a vascularized organ, bone is known to be susceptible to I/R injury too, but the respective mechanisms are not well understood to date. We therefore hypothesized that, similar to other organs, plasma cascade-induced inflammation also plays a role in bone I/R injury. Reperfusion injury in rat tibia was induced by unilateral clamping of the femoral artery and additional use of a tourniquet, while keeping the femoral vein patent to prevent venous congestion. Rats were subjected to 4h ischemia and 24h reperfusion. Deposition of complement fragment C3b/c and fibrin as well as expression of tissue factor (TF), tissue plasminogen activator (tPA), plasminogen activator inhibitor-1 (PAI-1), and E-selectin was detected by immunohistochemistry. In plasma, the levels of high mobility group box1 (HMGB1) were measured by ELISA. The total level of complement in serum was assessed by the CH50 test. Our results show that deposition of C3b/c was significantly increased with respect to healthy controls in cortical bone as well as in marrow of reperfused limbs. C3b/c deposition was also increased in cortical bone, but not in bone marrow, of contralateral limbs. Deposition of fibrin, as well as expression of PAI-1, was significantly increased in bone after ischemia and reperfusion, whereas expression of tPA was reduced. These differences were most prominent in vessels of bone, both in marrow and cortical bone, and both in reperfused and contralateral limbs. However, PAI-1, was only increased in vessels of reperfused cortical bone and there were no significant changes in expression of E-selectin. With respect to solid bone tissue, a significant increase of C3b/c and fibrin deposition was shown in osteocytes, and for fibrin also in the bone matrix, in both contralateral and reperfused cortical bone compared with normal healthy controls. A slight expression of TF was visible in osteocytes of the normal healthy control group, while TF was not present in the experimental groups. Moreover, CH50 values in serum decreased over time and HMGB1 was significantly increased in plasma of animals at the end of reperfusion. We conclude that ischemia and reperfusion of bone leads to activation of the complement and coagulation systems and a downregulation of the fibrinolytic cascade. In the acute phase, a vascular inflammation induced by activation of the plasma cascade systems also occurs in the bone. This is similar to I/R injury of other vascularized organs and tissues.


Subject(s)
Bone and Bones/pathology , Reperfusion Injury/blood , Animals , Bone Matrix/metabolism , Bone and Bones/blood supply , Bone and Bones/metabolism , Complement C3b/metabolism , E-Selectin/metabolism , Fibrin/metabolism , HMGB1 Protein/blood , Hindlimb/pathology , Male , Osteocytes/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Rats, Wistar , Reperfusion Injury/pathology , Sheep , Thromboplastin/metabolism , Tissue Plasminogen Activator/metabolism
9.
Am J Physiol Lung Cell Mol Physiol ; 312(1): L42-L55, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27815256

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a devastating lung disease with a median survival of 3 yr. IPF deteriorates upon viral or bacterial lung infection although pulmonary infection (pneumonia) in healthy lungs rarely induces fibrosis. Bacterial lipopolysaccharide (LPS) activates Toll-like receptor 4 (TLR4), initiating proinflammatory pathways. As TLR4 has already been linked to hepatic fibrosis and scleroderma, we now investigated the role of TLR4 in IPF fibroblasts. Lung tissue sections from patients with IPF were analyzed for TLR4 expression. Isolated normal human lung fibroblasts (NL-FB) and IPF fibroblasts (IPF-FB) were exposed to LPS and transforming growth factor-ß (TGF-ß) before expression analysis of receptors, profibrotic mediators, and cytokines. TLR4 is expressed in fibroblast foci of IPF lungs as well as in primary NL-FB and IPF-FB. As a model for a gram-negative pneumonia in the nonfibrotic lung, NL-FB and IPF-FB were coexposed to LPS and TGF-ß. Whereas NL-FB produced significantly less connective tissue growth factor upon costimulation compared with TGF-ß stimulation alone, IPF-FB showed significantly increased profibrotic markers compared with control fibroblasts after costimulation. Although levels of antifibrotic prostaglandin E2 were elevated after costimulation, they were not responsible for this effect. However, significant downregulation of TGF-ß receptor type 1 in control fibroblasts seems to contribute to the reduced profibrotic response in our in vitro model. Normal and IPF fibroblasts thus differ in their profibrotic response upon LPS-induced TLR4 stimulation.


Subject(s)
Fibroblasts/metabolism , Fibroblasts/pathology , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , Toll-Like Receptor 4/metabolism , Cell Separation , Cells, Cultured , Connective Tissue Growth Factor/metabolism , Dinoprostone/metabolism , ErbB Receptors/metabolism , Fibroblasts/drug effects , Humans , Lipopolysaccharides/pharmacology , Mutation/genetics , Smad3 Protein/metabolism , Toll-Like Receptor 4/genetics , Transforming Growth Factor beta/pharmacology
10.
Mol Hum Reprod ; 22(11): 768-777, 2016 11.
Article in English | MEDLINE | ID: mdl-27542948

ABSTRACT

STUDY QUESTION: Can the activity of the IκB kinase (IKKß) complex in endometriotic cells contribute to endometriotic lesion survival? SUMMARY ANSWER: There is a constitutive activity of the IKKß catalytic complex in peritoneal and deeply infiltrating lesions that can influence epithelial, but not stromal cell viability. WHAT IS KNOWN ALREADY: Endometriotic lesions exist in an inflammatory microenvironment with higher local concentrations of cytokines, such as tumour necrosis factor α (TNFα). TNFα stimulates the activation of the IKKß complex, an important nodal point in multiple signalling pathways that influence gene transcription, proliferation and apoptosis. However, few data on the regulation of IKKß in endometriotic tissue are currently available. STUDY DESIGN, SIZE, DURATION: A retrospective analysis of endometriotic tissue from peritoneal, ovarian and deeply infiltrating lesions from 37 women. PARTICIPANTS/MATERIALS, SETTING, METHODS: Basal and activated (phosphorylated) IKKß concentrations were analysed by western blotting and immunohistochemistry. The relationship between the expression and activation of these proteins and peritoneal fluid (TNFα) concentrations, measured via ELISA, was examined. A subsequent in vitro analysis of TNFα treatment on the activation of IKKß and the effect on epithelial and stromal cell viability by its inhibition with PS1145 was also performed. MAIN RESULTS AND ROLE OF CHANCE: Levels of the phosphorylated IKKß complex in endometriotic lesions had a significant positive correlation with peritoneal fluid TNFα concentrations. Phosphorylated IKKß complex was more prevalent in peritoneal and deeply infiltrating endometriosis lesions compared with ovarian lesions. IKKß was present in both epithelial and stromal cells in all lesions but active IKKß was limited to epithelial cells. TNFα stimulated an increased expression of phosphorylated IKKß and the inhibition of this kinase with PS1145 significantly influenced ectopic epithelial cells viability but not eutopic epithelial cells, or endometrial stromal cells. LIMITATIONS, REASONS FOR CAUTION: In vitro analysis on epithelial cells was performed with immortalized cell lines and not primary cell cultures and only low sample numbers were available for the study. WIDER IMPLICATIONS OF THE FINDINGS: The regulation of aberrant signalling pathways represents a promising yet relatively unexplored area of endometriosis progression. The IKKß complex is activated by inflammation and is critical nodal point of numerous downstream kinase-signalling pathways, including NFκB (nuclear factor κB), mTOR (mammalian target of rapamycin) and BAD (Bcl2-antagonist of cell death). This study shows a significant relationship between peritoneal fluid TNFα and IKKß activation in epithelial cells that will have significant consequences for the continued survival of these cells at ectopic locations through the regulation of downstream pathways. LARGE SCALE DATA: None. STUDY FUNDING/COMPETING INTERESTS: The study was funded by the Swiss National Science Foundation (Grant Number 320030_140774). The authors have no conflict of interest to declare.


Subject(s)
Endometriosis/metabolism , Endometriosis/pathology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , I-kappa B Kinase/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Transcription Factors/metabolism
11.
Dev Growth Differ ; 56(4): 255-75, 2014 May.
Article in English | MEDLINE | ID: mdl-24635767

ABSTRACT

Postnatally, the mammary gland undergoes continuous morphogenesis and thereby is especially prone to malignant transformation. Thus, the maintenance of the epithelium depends on a tight control of stem cell recruitment. We have previously shown that epithelial overexpression of the EphB4 receptor results in defective mammary epithelial development and conferred a metastasizing tumor phenotype on experimental mouse mammary tumors accompanied by a preponderance of progenitor cells. To analyze the effect of EphB4 overexpression on mammary epithelial cell fate, we have used Fluorescence Activated Cell Sorting (FACS) analyses to quantify epithelial sub-populations and repopulation assays of cleared fat pads to investigate their regenerative potential. These experiments revealed that deregulated EphB4 expression leads to an augmentation of bi-potent progenitor cells and to a shift of the differentiation pathway towards the luminal lineage. The analyses of the ductal outgrowths indicated that EphB4 overexpression leads to enforced branching activity, impedes ductal differentiation and stimulates angiogenesis. To elucidate the mechanisms forwarding EphB4 signals, we have compared the expression profile of defined cell populations between EphB4 transgene and wild type mammary glands concentrating on the wnt signaling pathway and on genes implicated in cell migration. With respect to wnt signaling, the progenitor cell population was the most affected, whereas the stem cell-enriched population showed the most pronounced deregulation of migration-associated genes. Thus, the luminal epithelial EphB4 signaling contributes, most likely via wnt signaling, to the regulation of migration and cell fate of early progenitors and is involved in the determination of branching points along the ductal tree.


Subject(s)
Cell Differentiation , Epithelium/metabolism , Mammary Glands, Animal/metabolism , Neovascularization, Physiologic/genetics , Receptor, EphB4/genetics , Receptor, EphB4/metabolism , Stem Cells/cytology , Animals , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Gene Expression Profiling , Mammary Glands, Animal/cytology , Mice , Mice, Transgenic , Polymerase Chain Reaction , Stem Cells/metabolism
12.
J Mol Endocrinol ; 52(2): 169-79, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24412827

ABSTRACT

Endometriosis is an extremely prevalent disorder characterized by the growth of endometrial tissue at ectopic locations. Glycolysis is an energy-producing mechanism that occurs in almost all cells and requires an adequate uptake of glucose mediated by glucose transporter (GLUT) proteins. At present, however, very little is known about their expression in either the endometrium or the endometriotic lesions. The objective of this study was to examine the expression of SLC2A genes in the endometrium of women with and without endometriosis and in the matching ectopic tissue, and to confirm the presence of the GLUT proteins in ectopic lesions. There was a significantly higher expression of SLC2A3 and a significantly lower expression of SLC2A4 in women with endometriosis compared with those without. In women with endometriosis, the ectopic expression of SLC2A3, SLC2A4 and SLC2A5 was significantly higher than that observed in the matching eutopic tissue. GLUT1 protein expression was present in both epithelial and stromal cells and GLUT3 was confined to CD45-positive leukocytes. GLUT4 expression was strong in both ectopic epithelial and stromal cells and localized to the cellular membrane in epithelial cells. These results show that GLUT expression is altered between eutopic and ectopic tissue and between women with and without endometriosis, and that GLUT4 may represent a significant entry route for glucose into the endometriotic epithelial cells. The inducible nature of GLUT4 and its limited cellular expression may make GLUT4 an attractive target for non-hormone-based treatments of endometriosis.


Subject(s)
Choristoma/genetics , Endometriosis/genetics , Endometrium/metabolism , Endometrium/pathology , Gene Expression Regulation , Glucose Transport Proteins, Facilitative/genetics , Adult , Cell Membrane/drug effects , Cell Membrane/metabolism , Choristoma/pathology , Endometriosis/pathology , Endometrium/blood supply , Endometrium/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Regulation/drug effects , Glucose Transport Proteins, Facilitative/metabolism , Hormones/pharmacology , Humans , Protein Transport/drug effects , Protein Transport/genetics
13.
Clin Cancer Res ; 19(1): 96-105, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23172887

ABSTRACT

PURPOSE: The phosphoinositide 3-kinase (PI3K) pathway is fundamental for cell proliferation and survival and is frequently altered and activated in neoplasia, including carcinomas of the lung. In this study, we investigated the potential of targeting the catalytic class I(A) PI3K isoforms in small cell lung cancer (SCLC), which is the most aggressive of all lung cancer types. EXPERIMENTAL DESIGN: The expression of PI3K isoforms in patient specimens was analyzed. The effects on SCLC cell survival and downstream signaling were determined following PI3K isoform inhibition by selective inhibitors or downregulation by siRNA. RESULTS: Overexpression of the PI3K isoforms p110-α and p110-ß and the antiapoptotic protein Bcl-2 was shown by immunohistochemistry in primary SCLC tissue samples. Targeting the PI3K p110-α with RNA interference or selective pharmacologic inhibitors resulted in strongly affected cell proliferation of SCLC cells in vitro and in vivo, whereas targeting p110-ß was less effective. Inhibition of p110-α also resulted in increased apoptosis and autophagy, which was accompanied by decreased phosphorylation of Akt and components of the mTOR pathway, such as the ribosomal S6 protein, and the eukaryotic translation initiation factor 4E-binding protein 1. A DNA microarray analysis revealed that p110-α inhibition profoundly affected the balance of pro- and antiapoptotic Bcl-2 family proteins. Finally, p110-α inhibition led to impaired SCLC tumor formation and vascularization in vivo. CONCLUSION: Together our data show the key involvement of the PI3K isoform p110-α in the regulation of multiple tumor-promoting processes in SCLC.


Subject(s)
Lung Neoplasms/enzymology , Phosphoinositide-3 Kinase Inhibitors , Small Cell Lung Carcinoma/enzymology , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival , Chick Embryo , Class Ia Phosphatidylinositol 3-Kinase/genetics , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Gene Expression Profiling , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Neovascularization, Pathologic/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , Signal Transduction , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , TOR Serine-Threonine Kinases/metabolism
14.
Fertil Steril ; 97(2): 373-80, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22154765

ABSTRACT

OBJECTIVE: To assess the relationship between endometriotic lesions with associated nerve fibers with both pain and peritoneal fluid (PF) cytokine concentrations based on lesion location. DESIGN: An observational study. SETTING: University hospital. PATIENT(S): Premenopausal women undergoing laparoscopy. INTERVENTION(S): The pain experienced by patients was recorded before surgery and ectopic endometrial tissue excised and matching PF collected during laparoscopy. Immunohistochemistry was performed on endometriotic tissue sections to identify nerve fibers and PF cytokine concentrations determined. MAIN OUTCOME MEASURE(S): The pain experienced by women with endometriosis, the lesion locations, and the prevalence and proximity of nerve fibers to endometriotic lesions, as well as the PF concentrations of multiple cytokines. RESULT(S): Lesions from the rectovaginal septum were significantly more likely to be associated with a nerve fiber and report more menstrual pain than lesions from other regions. The PF glycodelin concentrations were also significantly higher in samples with an endometriotic-associated nerve. In peritoneal endometriotic lesions significantly more menstrual pain was reported when endometriotic lesions were associated with nerve fibers, although no difference was observed between the cytokine concentrations. Ovarian endometriotic lesions were rarely associated with nerve fibers. CONCLUSION(S): The presence of endometriosis-associated nerve fibers appear to be related to both the pain experienced by women with endometriosis and the concentration of PF cytokines; however, this association varies with the lesion location.


Subject(s)
Cytokines/analysis , Endometriosis/complications , Nerve Fibers/pathology , Ovarian Diseases/complications , Pelvic Pain/etiology , Peritoneal Diseases/complications , Rectal Diseases/complications , Vaginal Diseases/complications , Analysis of Variance , Ascitic Fluid/immunology , Biomarkers/analysis , Endometriosis/immunology , Endometriosis/pathology , Endometriosis/surgery , Enzyme-Linked Immunosorbent Assay , Female , Hospitals, University , Humans , Immunohistochemistry , Laparoscopy , Nerve Fibers/chemistry , Ovarian Diseases/immunology , Ovarian Diseases/pathology , Ovarian Diseases/surgery , Pain Measurement , Pelvic Pain/immunology , Pelvic Pain/pathology , Peritoneal Diseases/immunology , Peritoneal Diseases/pathology , Peritoneal Diseases/surgery , Radioimmunoassay , Rectal Diseases/immunology , Rectal Diseases/pathology , Rectal Diseases/surgery , Switzerland , Ubiquitin Thiolesterase/analysis , Vaginal Diseases/immunology , Vaginal Diseases/pathology , Vaginal Diseases/surgery
15.
Int J Oncol ; 40(2): 357-69, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22020958

ABSTRACT

Cancer most probably originates from stem/progenitor cells and exhibits a similar cell hierarchy as normal tissues. Moreover, there is growing evidence that only the stem cells are capable of metastasis formation. We have previously shown that overexpression of a dominant negative ephrin-B2 mutant interferes with mammary gland differentiation and confers a metastatic phenotype to NeuT-induced mammary tumors with an increase in cells with stem/progenitor characteristics. To investigate the role of ephrin-B2 in the control of the mammary stem cell niche, we analyzed the mammary stem and progenitor cell populations in transgenic mice overexpressing the mutant ephrin-B2. Quantification by FACS analysis revealed a significant increase of cells in the basal/alveolar cell-, the bi-potent progenitor- and the stem cell-enriched fractions. Moreover, the supposed precursors of estrogen receptor-positive cells were elevated in the stem cell-enriched fraction. In contrast, the epithelium from transgenic mice overexpressing the native ephrin-B2 gene showed an augmentation of the luminal cell- and the bi-potent progenitor-enriched fractions. Repopulation assays revealed that the epithelial cells of truncated ephrin-B2 transgenic epithelial cells have a higher regeneration capacity than those of controls and of native ephrin-B2 transgenic mice, confirming the augmentation of stem cells. Morphologically, these outgrowths exhibited impaired basal/luminal compartmentalization and epithelial polarization. These results demonstrate that deregulated ephrin-B2 expression interferes with the regulation of the stem cell niche and leads to a shift of the differentiation pathway and may thereby contribute to the acquisition of the metastatic phenotype long before carcinogenic growth becomes apparent.


Subject(s)
Cell Differentiation , Ephrin-B2/metabolism , Epithelial Cells/metabolism , Mammary Glands, Animal/cytology , Stem Cell Niche , Stem Cells/physiology , Adaptor Proteins, Signal Transducing , Adipose Tissue/cytology , Animals , Cell Adhesion Molecules/metabolism , Cell Cycle Proteins , Cell Polarity , Cell Proliferation , Cell Transformation, Neoplastic , Ephrin-B2/genetics , Epithelial Cells/transplantation , Female , Homeostasis , Humans , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphoproteins/metabolism , Receptors, Estrogen/metabolism , Signal Transduction , Stem Cells/metabolism , Zonula Occludens-1 Protein
16.
Int J Oncol ; 38(1): 151-60, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21109936

ABSTRACT

We have previously shown that EphB4 and ephrin-B2 are differentially expressed in the mammary gland and that their deregulated expression in the mammary epithelium of transgenic mice leads to perturbations of the mammary parenchyma and vasculature. In addition, overexpression of EphB4 and expression of a truncated ephrin-B2 mutant, capable of receptor stimulation but incapable of reverse signalling, confers a metastasising phenotype on NeuT initiated mouse mammary tumours. We have taken advantage of this transgenic tumour model to compare stem cell characteristics between the non-metastasising and metastasising mammary tumours. We analysed the expression of the proliferation attenuating p21(waf) gene, which was significantly increased in the metastasising tumours. Moreover, we compared the expression of CK-19, Sca-1, CD24 and CD49f as markers for progenitor cells exhibiting a decreasing differentiation grade. Sca-1 expressing cells were the earliest progenitors detected in the non-metastasising NeuT induced tumours. The metastasising NeuT/EphB4 tumours were enriched in CD24 expressing cells, whereas the metastasising NeuT/truncated ephrin-B2 tumours contained in addition significant amounts of CD49f expressing cells. The same cell populations were also enriched in mammary glands of single transgenic MMTV-EphB4 and MMTV-truncated ephrin-B2 females indicating that deregulated EphB4-ephrin-B2 signalling interferes with the homeostasis of the stem/progenitor cell pool before tumour formation is initiated. Since the same cell populations are enriched in the normal tissue, primary mammary tumours and metastases we conclude that these progenitor cells were the origin of tumour formation and that this change in the tumour origin has led to the acquisition of the metastatic tumour phenotype.


Subject(s)
Ephrin-B2/biosynthesis , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptor, EphB4/biosynthesis , Animals , Cell Growth Processes/physiology , Ephrin-B2/antagonists & inhibitors , Ephrin-B2/genetics , Female , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Metastasis , Receptor, EphB4/genetics
17.
Am J Physiol Regul Integr Comp Physiol ; 299(2): R642-54, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20445153

ABSTRACT

Members of the ATP-binding cassette (ABC) transporters play a pivotal role in cellular lipid efflux. To identify candidate cholesterol transporters implicated in lipid homeostasis and mammary gland (MG) physiology, we compared expression and localization of ABCA1, ABCG1, and ABCA7 and their regulatory genes in mammary tissues of different species during the pregnancy-lactation cycle. Murine and bovine mammary glands (MGs) were investigated during different functional stages. The abundance of mRNAs was determined by quantitative RT-PCR. Furthermore, transporter proteins were localized in murine, bovine, and human MGs by immunohistochemistry. In the murine MG, ABCA1 mRNA abundance was elevated during nonlactating compared with lactating stages, whereas ABCA7 and ABCA1 mRNA profiles were not altered. In the bovine MG, ABCA1, ABCG1, and ABCA7 mRNAs abundances were increased during nonlactating stages compared with lactation. Furthermore, associations between mRNA levels of transporters and their regulatory genes LXRalpha, PPARgamma, and SREBPs were found. ABCA1, ABCG1, and ABCA7 proteins were localized in glandular MG epithelial cells (MEC) during lactation, whereas during nonlactating stages, depending on species, the proteins showed distinct localization patterns in MEC and adipocytes. Our results demonstrate that ABCA1, ABCG1, and ABCA7 are differentially expressed between lactation and nonlactating stages and in association with regulatory genes. Combined expression and localization data suggest that the selected cholesterol transporters are universal MG transporters involved in transport and storage of cholesterol and in lipid homeostasis of MEC. Because of the species-specific expression patterns of transporters in mammary tissue, mechanisms of cholesterol homeostasis seem to be differentially regulated between species.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cholesterol/metabolism , Lactation/metabolism , Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/genetics , Adipocytes/metabolism , Animals , Biological Transport , Cattle , Epithelial Cells/metabolism , Female , Gene Expression Regulation , Humans , Immunohistochemistry , Lipoproteins/metabolism , Liver X Receptors , Mice , Orphan Nuclear Receptors/genetics , PPAR gamma/genetics , Pregnancy , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity , Sterol Regulatory Element Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...