210 results on '"Lala PK"'
Search Results
2. Effects of colony stimulating factor-1 on human extravillous trophoblast growth and invasion
- Author
-
Hamilton, GS, primary, Lysiak, JJ, additional, Watson, AJ, additional, and Lala, PK, additional
- Published
- 1998
- Full Text
- View/download PDF
3. NG-nitro-L-arginine methyl ester, an inhibitor of nitric oxide synthesis, ameliorates interleukin 2-induced capillary leakage and reduces tumour growth in adenocarcinoma-bearing mice
- Author
-
Orucevic, A, primary and Lala, PK, additional
- Published
- 1996
- Full Text
- View/download PDF
4. Detection of antitrophoblast antibodies in the sera of patients with anticardiolipin antibodies and fetal loss
- Author
-
McCrae, KR, primary, DeMichele, AM, additional, Pandhi, P, additional, Balsai, MJ, additional, Samuels, P, additional, Graham, C, additional, Lala, PK, additional, and Cines, DB, additional
- Published
- 1993
- Full Text
- View/download PDF
5. Characterization of urokinase receptor expression by human placental trophoblasts
- Author
-
Zini, JM, primary, Murray, SC, additional, Graham, CH, additional, Lala, PK, additional, Kariko, K, additional, Barnathan, ES, additional, Mazar, A, additional, Henkin, J, additional, Cines, DB, additional, and McCrae, KR, additional
- Published
- 1992
- Full Text
- View/download PDF
6. Measurement of S period in growing cell populations by a graphic analysis of double labeling with 3H- and 14C-thymidine
- Author
-
Lala Pk
- Subjects
Double labeling ,Period (gene) ,Cell ,Cell Biology ,Biology ,Ehrlich ascites ,Molecular biology ,Graphic analysis ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,Immunology ,medicine ,Thymidine - Abstract
A graphic analysis is presented for the measurement of S period in asynchronously growing cell populations by double labeling with 3H and 14C- thymidine. It is applicable to growth characteristics anywhere between an exponential and a near steady state. Measurements made on Ehrlich ascites tumors of two different ages agree well with the values obtained by the labeled mitoses technique.
- Published
- 1968
- Full Text
- View/download PDF
7. Monoclonal origin of B lymphocyte colony-forming cells in spleen colonies formed by multipotential hemopoietic stem cells
- Author
-
Lala, PK and Johnson, GR
- Abstract
Spleen colonies produced by transplanting lethally irradiated mice with either 12 day fetal liver or adult bone marrow cells were found to contain B- lymphocyte colony-forming cells (BL-CFC) . The proportion of BL-CFC positive spleen colonies did not increase substantially between 8 and 14 days after transplantation, the range being 18-45 percent. However, the absolute number of BL-CFC per spleen colony varied considerably (between 1 and 10,318), although the majority of colonies contained less than 200 BL-CFC. Irrespective of the time after transplantation, smaller spleen colonies were found to have a higher frequency of BL-CFC than larger spleen colonies. To determine the possible clonal origin of BL-CFC from spleen colony- forming unit (CFU-S), CBA mice were injected with equal numbers of CBA and CBA T(6)/T(6) fetal liver or adult bone marrow cells. Analysis of 7-15-day spleen colonies demonstrated that 90 percent were either exclusively T(6) positive or T(6) negative and approximately equal numbers ofboth colony types were observed. B-lymphocyte colonies were grown and successfully karyotyped from 19 spleen colonies. When compared with the original spleen colony karyotype the B-lymphocyte colony cells karyotype was identical in all 19 cases. In 3 of the 19 colonies analyzed a mixture of T(6) positive and T(6) negative karyotypes was present and identical proportions of the karyotypes were present in the pooled B-lymphocyte colony cells and spleen colony cells. The data indicate that the B-lymphocyte colony-forming cells detected in spleen colonies are genuine members of the hemopoietic clone derived from the initiating hemopoietic stem cell (CFU-S).
- Published
- 1978
- Full Text
- View/download PDF
8. Testing using a minimal number of instructions
- Author
-
Lala, PK, primary
- Published
- 1981
- Full Text
- View/download PDF
9. Redefinable crossassembler for horizontally microprogrammable processors
- Author
-
Scholes, NF, primary, Morgan, G, additional, and Lala, PK, additional
- Published
- 1984
- Full Text
- View/download PDF
10. Editorial: Cellular and molecular determinants of pregnancy success at the fetal-maternal interface in health and disease.
- Author
-
Lala PK and Graham CH
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
11. Decorin-induced, preeclampsia-associated microRNA-512-3p restrains extravillous trophoblast functions by targeting USF2/PPP3R1 axis.
- Author
-
Halari CD, Nandi P, Sidhu J, Sbirnac M, Zheng M, and Lala PK
- Abstract
Decorin (DCN) is a leucine-rich proteoglycan produced by chorionic villus mesenchymal cells anddecidual cells during human pregnancy. Studies from our laboratory demonstrated that decidua-derived DCN restrains multiple trophoblast functions including proliferation, migration, invasion andendovascular differentiation, mediated by DCN-binding to multiple tyrosine kinase receptors; expressed by the trophoblast. Furthermore, DCN was shown to be selectively over-produced by thedecidua in preeclampsia (PE) subjects and elevated in the second trimester maternal plasma in PE, before the appearance of clinical signs, presenting as a predictive biomarker for PE. Micro (mi)RNAs are single-stranded non-coding RNAs (17-25 nucleotides) that typically downregulate target genes by repressing translation or facilitating degradation of mRNAs. The human; placenta expresses many miRNAs, some of which are exclusively expressed by the trophoblast. Many; of these miRNAs are dysregulated in PE-associated placentas and some appear in the maternal blood as PE biomarkers. However, little is known about their contribution to the pathogenesis of PE, a multi-factorial disease associated with a hypo-invasive placenta. The objective of the present study was to examine whether exposure of extravillous trophoblast (EVT) to DCN affects expression of specific miRNAs, and to test the role of these miRNAs in altering EVT functions. We identified miR-512-3p, as one of the DCN-induced miRNAs, also upregulated in PE placentas. It was shown to be elevated in ectopic DCN-over-expressing or exogenous DCN-treated first trimester human trophoblast cell line HTR-8/SVneo. Use of miRNA-mimics and inhibitors revealed that miR-512-3p compromised trophoblast migration, invasion and VEGF-dependent endovascular differentiation. Finally, Protein Phosphatase 3 Regulatory Subunit B, Alpha (PPP3R1), a known target of miR-512-3p, was paradoxically elevated in miR-512-3p-overexpressing trophoblast and PE-associated placentas. Using Enrichr, a tool that consists of both a validated user-submitted gene list and a search engine for transcription factors, we found that PPP3R1 elevation resulted from the miRNA binding to and targeting Upstream Transcription Factor 2 (USF2) which targeted PPP3R1. These findings reveal a novel aspect of pathogenesis of PE and biomarker potentials of this miRNA in PE., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Halari, Nandi, Sidhu, Sbirnac, Zheng and Lala.)
- Published
- 2022
- Full Text
- View/download PDF
12. A crossroad between placental and tumor biology: What have we learnt?
- Author
-
Lala PK, Nandi P, Hadi A, and Halari C
- Subjects
- Choriocarcinoma pathology, Female, Humans, Pregnancy, Uterine Neoplasms pathology, Neoplasms pathology, Placenta pathology, Placentation physiology
- Abstract
Placenta in certain species including the human has evolved as a highly invasive tumor-like organ invading the uterus aned its vasculature to derive oxygen and nutrients for the fetus and exchange waste products. While several excellent reviews have been written comparing hemochorial placentation with tumors, no comprehensive review is available dealing with mechanistic insights into what makes them different, and what tumor biologists can learn from placental biologists, and vice versa. In this review, we analyze the structure-function relationship of the human placenta, emphasizing the functional need of the spatio-temporally orchestrated trophoblast invasiveness for fetal development and growth, and pathological consequences of aberrant invasiveness for fetal and maternal health. We then analyze similarities and differences between the placenta and invasive tumors in terms of hallmarks of cancer, some key molecules regulating their invasive functions, and how placental cancers (choriocarcinomas) or other cancers become refractory or even addicted to these invasion-restraining molecules. We cite in vitro models of human trophoblast and choriocarcinoma cell lines utilized to study mechanisms in normal placental development as well as those responsible for tumor progression. We discuss the pathobiology of hyper-invasive placentas and show thattrophoblastic neoplasias are a unique and heterogeneous class of tumors. We delve into the questions as to why metastasis from other organs rarely occurs at the placental site and whether pregnancy makes the mother more or less vulnerable to cancer-related morbidity/mortality. We attempt to compare trophoblast stem cells and cancer stem cells. Finally, we leave the readers with some thoughts as foods of future investigations., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
13. Molecular mechanisms in IL-1β-mediated decorin production by decidual cells.
- Author
-
Halari CD, Renaud SJ, and Lala PK
- Subjects
- Active Transport, Cell Nucleus, Binding Sites, Cell Line, Decidua metabolism, Decorin genetics, Female, Humans, Interleukin-1beta metabolism, Macrophages metabolism, NF-kappa B metabolism, Pregnancy, Pregnancy Trimester, First, Promoter Regions, Genetic, Receptors, Interleukin-1 Type I metabolism, Stromal Cells metabolism, Up-Regulation, Decidua drug effects, Decorin metabolism, Interleukin-1beta pharmacology, Macrophages drug effects, Receptors, Interleukin-1 Type I agonists, Stromal Cells drug effects
- Abstract
Decorin, a small leucine-rich proteoglycan produced by decidual cells restrains trophoblast differentiation, migration and invasiveness of extra-villous trophoblast cells. Decidual overproduction of decorin is associated with preeclampsia, and elevated decorin levels in maternal plasma are a predictive biomarker of preeclampsia. Furthermore, decorin plays an autocrine role in maturation of human endometrial stromal cells into decidual cells. Thus, a balanced decorin production by the decidua is critical for healthy pregnancy. However, the molecular mechanisms regulating decorin production by the decidua are unclear. Interleukin-1 beta is an inflammation-associated multi-functional cytokine, and is reported to induce decidualization in primates. Hence, the present study was designed: (i) to test if exogenous Interleukin-1 beta stimulated decorin production by human endometrial stromal cells; and if so, (ii) to identify the cellular source of Interleukin-1 beta in first trimester decidual tissue; (iii) to identify the downstream molecular partners in Interleukin-1 beta mediated decorin production by human endometrial stromal cells. Results revealed that (i) amongst multiple pro-inflammatory cytokines tested, Interleukin-1 beta alone stimulated decorin production by these cells; (ii) both macrophages and decidual cells in first trimester decidua produced Interleukin-1 beta; (iii) Interleukin-1 beta mediated decorin production was dependent on Interleukin-1 receptor activation, followed by activation and nuclear translocation of nuclear factor kappa B and its binding to the decorin promoter. These results reveal that Interleukin-1 beta plays a novel role in inducing decorin production by human endometrial stromal cells by activating nuclear factor kappa B., (© The Author(s) 2021. Published by Oxford University Press on behalf of European Society of Human Reproduction and Embryology. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
14. Roles of Two Small Leucine-Rich Proteoglycans Decorin and Biglycan in Pregnancy and Pregnancy-Associated Diseases.
- Author
-
Halari CD, Zheng M, and Lala PK
- Subjects
- Animals, Biglycan metabolism, Decorin metabolism, Female, Fetal Growth Retardation metabolism, Gene Expression Regulation, Humans, Mutation, Placenta metabolism, Pre-Eclampsia metabolism, Pregnancy, Biglycan genetics, Decorin genetics, Fetal Growth Retardation genetics, Pre-Eclampsia genetics
- Abstract
Two small leucine-rich proteoglycans (SLRP), decorin and biglycan, play important roles in structural-functional integrity of the placenta and fetal membranes, and their alterations can result in several pregnancy-associated diseases. In this review, we briefly discuss normal placental structure and functions, define and classify SLRPs, and then focus on two SLRPs, decorin (DCN) and biglycan (BGN). We discuss the consequences of deletions/mutations of DCN and BGN. We then summarize DCN and BGN expression in the pregnant uterus, myometrium, decidua, placenta, and fetal membranes. Actions of these SLRPs as ligands are then discussed in the context of multiple binding partners in the extracellular matrix and cell surface (receptors), as well as their alterations in pathological pregnancies, such as preeclampsia, fetal growth restriction, and preterm premature rupture of membranes. Lastly, we raise some unanswered questions as food for thought.
- Published
- 2021
- Full Text
- View/download PDF
15. Pri-miR526b and Pri-miR655 Are Potential Blood Biomarkers for Breast Cancer.
- Author
-
Majumder M, Ugwuagbo KC, Maiti S, Lala PK, and Brackstone M
- Abstract
We reported that two microRNAs, miR526b and miR655, are oncogenic in breast cancer (BC). Overexpression of these two miRNAs in poorly metastatic BC cells promotes aggressive BC phenotypes in vitro and in vivo. High expression of each miRNA was associated with poor patient survival. In this pilot biomarker study, we report for the first time that miRNA precursor RNAs (pri-miRNAs) are robust and sensitive biomarkers for BC, detectable in both human blood plasma and biopsy tissues. Pri-miRNA detection and quantification do not require a special enrichment procedure, thus reducing specimen quantity. Blood plasma samples from 90 malignant tumor-bearing patients and 20 benign lesion-bearing participants (control) were analyzed for pri-miRNA expression with a quantitative real-time polymerase chain reaction. Results revealed that normalized expressions of plasma pri-miR526b and pri-miR655 are significantly upregulated in malignancy compared to benign plasmas ( p = 0.002 and p = 0.03, respectively). Both pri-miRNAs showed more prominent results to distinguish stage I plasmas from benign plasmas ( p = 0.001 for pri-miR526b and p = 0.0001 for pri-miR655). We have also validated pri-miRNA expression in independent tumor bank tissues, showing significant upregulation of both pri-miRNAs in BC; thus, pri-miRNAs are robust markers. The diagnostic relevance of pri-miRNAs was computed with the area under the curve (AUC). Pri-miR526b is a sensitive biomarker to distinguish cancer from control plasmas (sensitivity of 86%; AUC = 71.47%, p = 0.0027) with a positive predictive value of 88.89%; however, pri-miR655 did not show significant sensitivity. Furthermore, pri-miR526b could also significantly distinguish tumors as early as stage I from control (sensitivity of 75%; AUC = 72.71%, p = 0.0037). Therefore, pri-miR526b can be used as an early diagnostic biomarker. The expression of both pri-miRNAs was significantly high in ER-positive and HER2-negative subgroups of BC; hence, these biomarkers might play a role in the management of endocrine therapy designs. Additionally, with a case-control cohort study, we identified that high expression of pri-miR526b in the blood is also a risk factor associated with breast cancer (OR = 4.3, CI = 1.39-13.34, p = 0.01). Pri-miRNAs could be considered novel breast cancer blood biomarkers.
- Published
- 2021
- Full Text
- View/download PDF
16. Prostaglandin E2 Receptor 4 (EP4) as a Therapeutic Target to Impede Breast Cancer-Associated Angiogenesis and Lymphangiogenesis.
- Author
-
De Paz Linares GA, Opperman RM, Majumder M, and Lala PK
- Abstract
The formation of new blood (angiogenesis) and lymphatic (lymphangiogenesis) vessels are major events associated with most epithelial malignancies, including breast cancer. Angiogenesis is essential for cancer cell survival. Lymphangiogenesis is critical in maintaining tumoral interstitial fluid balance and importing tumor-facilitatory immune cells. Both vascular routes also serve as conduits for cancer metastasis. Intratumoral hypoxia promotes both events by stimulating multiple angiogenic/lymphangiogenic growth factors. Studies on tumor-associated lymphangiogenesis and its exploitation for therapy have received less attention from the research community than those on angiogenesis. Inflammation is a key mediator of both processes, hijacked by many cancers by the aberrant expression of the inflammation-associated enzyme cyclo-oxygenase (COX)-2. In this review, we focus on breast cancer and showed that COX-2 is a major promoter of both events, primarily resulting from the activation of prostaglandin (PG) E receptor EP4 on tumor cells, tumor-infiltrating immune cells, and endothelial cells; and the induction of oncogenic microRNAs. The COX-2/EP4 pathway also promotes additional events in breast cancer progression, such as cancer cell migration, invasion, and the stimulation of stem-like cells. Based on a combination of studies using multiple breast cancer models, we show that EP4 antagonists hold a major promise in breast cancer therapy in combination with other modalities including immune check-point inhibitors.
- Published
- 2021
- Full Text
- View/download PDF
17. Decorin production by the human decidua: role in decidual cell maturation.
- Author
-
Halari CD, Nandi P, Jeyarajah MJ, Renaud SJ, and Lala PK
- Subjects
- Cells, Cultured, Endometrium metabolism, Female, Gestational Age, HEK293 Cells, Humans, Placenta metabolism, Pregnancy, Trophoblasts metabolism, Decidua metabolism, Decorin metabolism, Embryo Implantation physiology
- Abstract
Decidualization involves the proliferation and differentiation of fibroblast-like endometrial stromal cells into epithelioid-shaped and secretory 'decidual' cells in response to steroid hormones. Human decidual cells produce insulin-like growth factor-binding protein-1 and prolactin (PRL), two well-recognized markers of decidual cell maturation and a proteoglycan decorin (DCN). We reported that DCN restrains the human trophoblast renewal, migration, invasion and endovascular differentiation needed for uterine arterial remodeling during normal pregnancy. DCN overproduction by the decidua is associated with a hypo-invasive placenta and a serious pregnancy disorder, pre-eclampsia (PE). Furthermore, elevated maternal plasma DCN levels during the second trimester is a predictive biomarker of PE. While these paracrine roles of decidua-derived DCN on trophoblast physiology and pathology have been well-defined, it remains unknown whether DCN plays any autocrine role in decidual cell development. The objectives of this study were to examine: the kinetics of DCN production during decidualization of human endometrial stromal cells; gestational age-related changes in DCN production by the first trimester decidua; and a possible autocrine role of DCN on decidual cell maturation. We found that DCN production is enhanced during decidualization of both primary and immortalized human endometrial stromal cells in vitro and during early gestation in decidual samples tested ex vivo, and that it is important for endometrial stromal cell maturation into a decidual phenotype. Decorin-depleted human endometrial stromal cells exposed to decidualizing stimuli failed to mature fully, as evidenced by fibroblastoid morphology, reduced insulin-like growth factor-binding protein-1 and PRL expression, and reduction in cellular ploidy. We identified heart and neural crest derivatives-expressed protein 2, and progesterone receptor as potential downstream mediators of DCN effects., (© The Author(s) 2020. Published by Oxford University Press on behalf of European Society of Human Reproduction and Embryology. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
18. Tumor suppressor role of cytoplasmic polyadenylation element binding protein 2 (CPEB2) in human mammary epithelial cells.
- Author
-
Tordjman J, Majumder M, Amiri M, Hasan A, Hess D, and Lala PK
- Subjects
- Animals, CRISPR-Cas Systems, Cell Movement, Cell Proliferation, Cyclooxygenase 2 metabolism, Epithelial Cells metabolism, Epithelial-Mesenchymal Transition, Female, Gene Knockout Techniques, Heterografts, Humans, MCF-7 Cells, Mice, Mice, Inbred NOD, Mice, SCID, MicroRNAs metabolism, Protein Isoforms, RNA, Small Interfering metabolism, RNA-Binding Proteins genetics, Tumor Suppressor Protein p53 metabolism, Breast cytology, Breast Neoplasms pathology, RNA-Binding Proteins metabolism
- Abstract
Background: Over-expression of cyclooxygenase (COX)-2 promotes breast cancer progression by multiple mechanisms, including induction of stem-like cells (SLC). Combined gene expression and microRNA microarray analyses of empty vector vs COX-2- transfected COX-2 low MCF7 breast cancer cell line identified two COX-2-upregulated microRNAs, miR-526b and miR-655, both found to be oncogenic and SLC-promoting. Cytoplasmic Polyadenylation Element-Binding Protein 2 (CPEB2) was the single common target of both microRNAs, the functions of which remain controversial. CPEB2 has multiple isoforms (A-F), and paradoxically, a high B/A ratio was reported to impart anoikis-resistance and metastatic phenotype in triple- negative breast cancer cells. We tested whether CPEB2 is a tumor suppressor in mammary epithelial cells., Methods: We knocked-out CPEB2 in the non-tumorigenic mammary epithelial cell line MCF10A by CRISPR/Cas9-double nickase approach, and knocked-down CPEB2 with siRNAs in the poorly malignant MCF7 cell line, both lines being high CPEB2-expressing. The resultant phenotypes for oncogenity were tested in vitro for both lines and in vivo for CPEB2KO cells. Finally, CPEB2 expression was compared between human breast cancer and non-tumor breast tissues., Results: CPEB2 (isoform A) expression was inversely correlated with COX-2 or the above microRNAs in COX-2-divergent breast cancer cell lines. CPEB2KO MCF10A cells exhibited oncogenic properties including increased proliferation, migration, invasion, EMT (decreased E-Cadherin, increased Vimentin, N-Cadherin, SNAI1, and ZEB1) and SLC phenotype (increased tumorsphere formation and SLC marker-expression). Tumor-suppressor p53 protein was shown to be a novel translationally-regulated target of CPEB2, validated with polysome profiling. CPEB2KO, but not wild-type cells produced lung colonies upon intravenous injection and subcutaneous tumors and spontaneous lung metastases upon implantation at mammary sites in NOD/SCID/IL2Rϒ-null mice, identified with HLA immunostaining. Similarly, siRNA-mediated CPEB2 knockdown in MCF7 cells promoted oncogenic properties in vitro. Human breast cancer tissues (n = 105) revealed a lower mRNA expression for CPEB2 isoform A and also a lower A/B isoform ratio than in non-tumour breast tissues (n = 20), suggesting that CPEB2A accounts for the tumor-suppressor functions of CPEB2., Conclusions: CPEB2, presumably the isoform A, plays a key role in suppressing tumorigenesis in mammary epithelial cells by repressing EMT, migration, invasion, proliferation and SLC phenotype, via multiple targets, including a newly-identified translational target p53.
- Published
- 2019
- Full Text
- View/download PDF
19. Roles of prostaglandins in tumor-associated lymphangiogenesis with special reference to breast cancer.
- Author
-
Lala PK, Nandi P, and Majumder M
- Subjects
- Animals, Biomarkers, Breast Neoplasms complications, Cyclooxygenase 2 metabolism, Disease Progression, Eicosanoids metabolism, Endothelial Cells metabolism, Female, Humans, In Vitro Techniques, Lymphatic Metastasis, Lymphedema etiology, Metabolic Networks and Pathways, Receptors, Prostaglandin metabolism, Signal Transduction, Breast Neoplasms metabolism, Breast Neoplasms pathology, Lymphangiogenesis, Neovascularization, Pathologic, Prostaglandins metabolism, Tumor Microenvironment
- Abstract
Lymphangiogenesis (formation of new lymphatic vessels), unlike angiogenesis, has been a lesser-focused field in cancer biology, because of earlier controversy regarding whether lymphatic metastasis occurs via pre-existing or newly formed lymphatics. Recent evidence reveals that peri-tumoral or intra-tumoral lymphangiogenesis is a precursor for lymphatic metastasis in most carcinomas and melanomas. Two major lymphangiogenic factors, vascular endothelial growth factor (VEGF)-C and VEGF-D, are produced by cancer cells or immune cells such as macrophages in the tumor-stroma to promote sprouting of lymphatics from lymphatic endothelial cells (LEC) or LEC precursors (LECP) by binding to their primary (high affinity) receptor VEGF-R3 or secondary receptors VEGF-R2, neuropilin (NRP)2 and α9/β1 integrin. Many other growth factors/receptors such as VEGF-A/VEGF-R2, fibroblast growth factor (FGF)2/FGF-R, platelet-derived growth factor (PDGF)/PDGF-R, hepatocyte growth factor (HGF)/C-Met, angiopoietins (Ang)1, 2/Tie2, and chemokines/ chemokine receptors (CCL21/CCR7, CCL12/CCR4) can also stimulate LEC sprouting directly or indirectly. This review deals with the roles of prostaglandins (PG), in particular PGE2, in cancer-associated lymphangiogenesis, with special emphasis on breast cancer. We show that cyclooxygenase (COX)-2 expression by breast cancer cells or tumor stroma leading to high PGE2 levels in the tumor milieu promotes lymphangiogenesis and lymphatic metastases, resulting from binding of PGE2 to PGE receptors (EP, in particular EP4) on multiple cell types: tumor cells, tumor-infiltrating immune cells, and LEC. EP4 activation on cancer cells and macrophages upregulated VEGF-C/D production to stimulate LEC sprouting. Furthermore, ligation of EP4 with PGE2 on cancer or host cells can initiate a new cascade of molecular events leading to cross-talk between cancer cells and LEC, facilitating lymphangiogenesis and lympho-vascular transport of cancer cells. We make a case for EP4 as a potential therapeutic target for breast cancer.
- Published
- 2018
- Full Text
- View/download PDF
20. Human trophoblast stem cell self-renewal and differentiation: Role of decorin.
- Author
-
Nandi P, Lim H, Torres-Garcia EJ, and Lala PK
- Subjects
- Cell Line, Transformed, Female, Human Embryonic Stem Cells cytology, Humans, Pregnancy, Pregnancy Trimester, First, Trophoblasts cytology, Antigens, Differentiation metabolism, Cell Differentiation, Decorin metabolism, Human Embryonic Stem Cells metabolism, Trophoblasts metabolism
- Abstract
The origin and regulation of stem cells sustaining trophoblast renewal in the human placenta remain unclear. Decorin, a leucine-rich proteoglycan restrains trophoblast proliferation, migration/invasiveness and endovascular differentiation, and local decorin overproduction is associated with preeclampsia (PE). Here, we tested the role of decorin in human trophoblast stem cell self-renewal and differentiation, using two models: an immortalized first trimester trophoblast cell line HTR-8/SVneo (HTR) and freshly isolated primary trophoblast (p-trophoblast) from early first trimester (6-9 weeks) placentas. Self-renewal capacity was measured by spheroid forming ability of single cells on ultra-low attachment plates for multiple generations. Markers of embryonic stem (ES) cells, trophoblast stem (TS) cells and trophoblast were used to identify stem cell hierarchy. Differentiation markers for syncytial and extravillous (EVT) pathways were employed to identify differentiated cells. Bewo cells were additionally used to explore DCN effects on syncytialization. Results reveal that the incidence of spheroid forming stem-like cells was 13-15% in HTR and 0.1-0.4%, in early first trimester p-trophoblast, including a stem cell hierarchy of two populations of ES and TS-like cells. DCN restrained ES cell self-renewal, promoted ES to TS transition and maintenance of TS cell stem-ness, but inhibited TS cell differentiation into both syncytial and EVT pathways.
- Published
- 2018
- Full Text
- View/download PDF
21. EP4 as a Therapeutic Target for Aggressive Human Breast Cancer.
- Author
-
Majumder M, Nandi P, Omar A, Ugwuagbo KC, and Lala PK
- Subjects
- Dinoprostone genetics, Dinoprostone metabolism, Female, Humans, MicroRNAs genetics, MicroRNAs metabolism, Neoplasm Invasiveness, Neoplasm Metastasis, RNA, Neoplasm genetics, RNA, Neoplasm metabolism, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Receptors, Prostaglandin E, EP4 Subtype antagonists & inhibitors, Receptors, Prostaglandin E, EP4 Subtype genetics, Receptors, Prostaglandin E, EP4 Subtype metabolism, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms metabolism, Triple Negative Breast Neoplasms pathology
- Abstract
G-protein-coupled receptors (GPCRs, also called seven-transmembrane or heptahelical receptors) are a superfamily of cell surface receptor proteins that bind to many extracellular ligands and transmit signals to an intracellular guanine nucleotide-binding protein (G-protein). When a ligand binds, the receptor activates the attached G-protein by causing the exchange of Guanosine-5'-triphosphate (GTP) for guanosine diphosphate (GDP). They play a major role in many physiological functions, as well as in the pathology of many diseases, including cancer progression and metastasis. Only a few GPCR members have been exploited as targets for developing drugs with therapeutic benefit in cancer. Present review briefly summarizes the signaling pathways utilized by the EP (prostaglandin E receptor) family of GPCR, their physiological and pathological roles in carcinogenesis, with special emphasis on the roles of EP4 in breast cancer progression. We make a case for EP4 as a promising newer therapeutic target for treating breast cancer. We show that an aberrant over-expression of cyclooxygenase (COX)-2, which is an inflammation-associated enzyme, occurring in 40-50% of breast cancer patients leads to tumor progression and metastasis due to multiple cellular events resulting from an increased prostaglandin (PG) E2 production in the tumor milieu. They include inactivation of host anti-tumor immune cells, such as Natural Killer (NK) and T cells, increased immuno-suppressor function of tumor-associated macrophages, promotion of tumor cell migration, invasiveness and tumor-associated angiogenesis, due to upregulation of multiple angiogenic factors including Vascular Endothelial Growth Factor (VEGF)-A, increased lymphangiogenesis (due to upregulation of VEGF-C/D), and a stimulation of stem-like cell (SLC) phenotype in cancer cells. All of these events were primarily mediated by activation of the Prostaglandin (PG) E receptor EP4 on tumor or host cells. We show that selective EP4 antagonists (EP4A) could mitigate all of these events tested with cells in vitro as well as in vivo in syngeneic COX-2 expressing mammary cancer bearing mice or immune-deficient mice bearing COX-2 over-expressing human breast cancer xenografts. We suggest that EP4A can avoid thrombo-embolic side effects of long term use of COX-2 inhibitors by sparing cardio-protective roles of PGI2 via IP receptor activation or PGE2 via EP3 receptor activation. Furthermore, we identified two COX-2/EP4 induced oncogenic and SLC-stimulating microRNAs-miR526b and miR655, one of which (miR655) appears to be a potential blood biomarker in breast cancer patients for monitoring SLC-ablative therapies, such as with EP4A. We suggest that EP4A will likely produce the highest benefit in aggressive breast cancers, such as COX-2 expressing triple-negative breast cancers, when combined with other newer agents, such as inhibitors of programmed cell death (PD)-1 or PD-L1., Competing Interests: The authors declare no conflict of interest.
- Published
- 2018
- Full Text
- View/download PDF
22. COX-2 induces oncogenic micro RNA miR655 in human breast cancer.
- Author
-
Majumder M, Dunn L, Liu L, Hasan A, Vincent K, Brackstone M, Hess D, and Lala PK
- Subjects
- Animals, Breast Neoplasms mortality, Breast Neoplasms pathology, Cell Line, Tumor, Cell Movement genetics, Cell Proliferation genetics, Cyclooxygenase 2 genetics, Cyclooxygenase 2 Inhibitors pharmacology, Dinoprostone metabolism, Disease Models, Animal, Female, Gene Knockdown Techniques, Humans, Lung Neoplasms pathology, Lung Neoplasms secondary, MCF-7 Cells, Mice, NF-kappa B metabolism, Neoplastic Stem Cells metabolism, Phenotype, Phosphorylation, Prognosis, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Proto-Oncogene Proteins c-akt metabolism, RNA Interference, Receptors, Prostaglandin E, EP4 Subtype genetics, Receptors, Prostaglandin E, EP4 Subtype metabolism, Signal Transduction drug effects, Smad3 Protein metabolism, Transforming Growth Factor beta metabolism, Breast Neoplasms genetics, Breast Neoplasms metabolism, Cyclooxygenase 2 metabolism, Gene Expression Regulation, Neoplastic drug effects, MicroRNAs genetics
- Abstract
We show that Cyclooxygenase-2 over-expression induces an oncogenic microRNA miR655 in human breast cancer cells by activation of EP4. MiR655 expression positively correlated with COX-2 in genetically disparate breast cancer cell lines and increased in all cell lines when grown as spheroids, implicating its link with stem-like cells (SLCs). Ectopic miR655 over-expression in MCF7 and SKBR3 cells resulted in increased proliferation, migration, invasion, spheroid formation and Epithelial to Masenchymal transition (EMT). Conversely, knocking down miR655 in aggressive MCF7-COX2 and SKBR3-COX2 cells reverted these phenotypes. MCF7-miR655 cells displayed upregulated NOTCH/WNT genes; both pathway inhibitors abrogated miR655-induced spheroid formation, linking miR655 with SLC-related pathways. MiR655 expression was dependent on EP4 activity and EP4 downstream signaling pathways PI3K/AKT, ERK and NF-kB and led to TGFβ resistance for Smad3 phosphorylation. Tail vein injection of MCF7-miR655 and SKBR3-miR655 cells in NOD/SCID/GUSB-null mice revealed increased lung colony growth and micrometastases to liver and spleen. MiR655 expression was significantly high in human breast tumors (n = 105) compared to non-tumor tissues (n = 20) and associated with reduced patient survival. Thus miR655 could serve as a prognostic breast cancer biomarker.
- Published
- 2018
- Full Text
- View/download PDF
23. PGE2 promotes breast cancer-associated lymphangiogenesis by activation of EP4 receptor on lymphatic endothelial cells.
- Author
-
Nandi P, Girish GV, Majumder M, Xin X, Tutunea-Fatan E, and Lala PK
- Subjects
- Animals, Blotting, Western, Cell Line, Tumor, Endothelial Cells pathology, Enzyme-Linked Immunosorbent Assay, Female, Mice, Mice, Nude, Rats, Real-Time Polymerase Chain Reaction, Dinoprostone metabolism, Endothelial Cells metabolism, Lymphangiogenesis physiology, Mammary Neoplasms, Experimental pathology, Receptors, Prostaglandin E, EP4 Subtype metabolism
- Abstract
Background: Lymphatic metastasis, facilitated by lymphangiogenesis is a common occurrence in breast cancer, the molecular mechanisms remaining incompletely understood. We had earlier shown that cyclooxygenase (COX)-2 expression by human or murine breast cancer cells promoted lymphangiogenesis and lymphatic metastasis by upregulating VEGF-C/D production by tumor cells or tumor-associated macrophages primarily due to activation of the prostaglandin receptor EP4 by endogenous PGE2. It is not clear whether tumor or host-derived PGE2 has any direct effect on lymphangiogenesis, and if so, whether EP4 receptors on lymphatic endothelial cells (LEC) play any role., Methods: Here, we address these questions employing in vitro studies with a COX-2-expressing and VEGF-C/D-producing murine breast cancer cell line C3L5 and a rat mesenteric (RM) LEC line and in vivo studies in nude mice., Results: RMLEC responded to PGE2, an EP4 agonist PGE1OH, or C3L5 cell-conditioned media (C3L5-CM) by increased proliferation, migration and accelerated tube formation on growth factor reduced Matrigel. Native tube formation by RMLEC on Matrigel was abrogated in the presence of a selective COX-2 inhibitor or an EP4 antagonist. Addition of PGE2 or EP4 agonist, or C3L5-CM individually in the presence of COX-2 inhibitor, or EP4 antagonist, restored tube formation, reinforcing the role of EP4 on RMLEC in tubulogenesis. These results were partially duplicated with a human dermal LEC (HMVEC-dLyAd) and a COX-2 expressing human breast cancer cell line MDA-MB-231. Knocking down EP4 with shRNA in RMLEC abrogated their tube forming capacity on Matrigel in the absence or presence of PGE2, EP4 agonist, or C3L5-CM. RMLEC tubulogenesis following EP4 activation by agonist treatment was dependent on PI3K/Akt and Erk signaling pathways and VEGFR-3 stimulation. Finally in a directed in vivo lymphangiogenesis assay (DIVLA) we demonstrated the lymphangiogenic as well as angiogenic capacity of PGE2 and EP4 agonist in vivo., Discussion/conclusions: These results demonstrate the roles of tumor as well as host-derived PGE2 in inducing lymphangiogenesis, at least in part, by activating EP4 and VEGFR-3 on LEC. EP4 being a common target on both tumor and host cells contributing to tumor-associated lymphangiogenesis reaffirms the therapeutic value of EP4 antagonists in the intervention of lymphatic metastasis in breast cancer.
- Published
- 2017
- Full Text
- View/download PDF
24. COX-2 Induces Breast Cancer Stem Cells via EP4/PI3K/AKT/NOTCH/WNT Axis.
- Author
-
Majumder M, Xin X, Liu L, Tutunea-Fatan E, Rodriguez-Torres M, Vincent K, Postovit LM, Hess D, and Lala PK
- Subjects
- Animals, Breast Neoplasms genetics, Cell Line, Tumor, Female, Gene Expression Regulation, Neoplastic, Humans, Mice, Neoplasm Invasiveness, Neoplasm Micrometastasis pathology, Phenotype, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Receptors, Notch metabolism, Receptors, Prostaglandin E, EP4 Subtype metabolism, Wnt Proteins metabolism, Breast Neoplasms enzymology, Breast Neoplasms pathology, Cyclooxygenase 2 metabolism, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Signal Transduction
- Abstract
Cancer stem-like cells (SLC) resist conventional therapies, necessitating searches for SLC-specific targets. We established that cyclo-oxygenase(COX)-2 expression promotes human breast cancer progression by activation of the prostaglandin(PG)E-2 receptor EP4. Present study revealed that COX-2 induces SLCs by EP4-mediated NOTCH/WNT signaling. Ectopic COX-2 over-expression in MCF-7 and SKBR-3 cell lines resulted in: increased migration/invasion/proliferation, epithelial-mesenchymal transition (EMT), elevated SLCs (spheroid formation), increased ALDH activity and colocalization of COX-2 and SLC markers (ALDH1A, CD44, β-Catenin, NANOG, OCT3/4, SOX-2) in spheroids. These changes were reversed with COX-2-inhibitor or EP4-antagonist (EP4A), indicating dependence on COX-2/EP4 activities. COX-2 over-expression or EP4-agonist treatments of COX-2-low cells caused up-regulation of NOTCH/WNT genes, blocked with PI3K/AKT inhibitors. NOTCH/WNT inhibitors also blocked COX-2/EP4 induced SLC induction. Microarray analysis showed up-regulation of numerous SLC-regulatory and EMT-associated genes. MCF-7-COX-2 cells showed increased mammary tumorigenicity and spontaneous multiorgan metastases in NOD/SCID/IL-2Rγ-null mice for successive generations with limiting cell inocula. These tumors showed up-regulation of VEGF-A/C/D, Vimentin and phospho-AKT, down-regulation of E-Cadherin and enrichment of SLC marker positive and spheroid forming cells. MCF-7-COX-2 cells also showed increased lung colonization in NOD/SCID/GUSB-null mice, an effect reversed with EP4-knockdown or EP4A treatment of the MCF-7-COX-2 cells. COX-2/EP4/ALDH1A mRNA expression in human breast cancer tissues were highly correlated with one other, more marked in progressive stage of disease. In situ immunostaining of human breast tumor tissues revealed co-localization of SLC markers with COX-2, supporting COX-2 inducing SLCs. High COX-2/EP4 mRNA expression was linked with reduced survival. Thus, EP4 represents a novel SLC-ablative target in human breast cancer. Stem Cells 2016;34:2290-2305., (© 2016 AlphaMed Press.)
- Published
- 2016
- Full Text
- View/download PDF
25. Decorin over-expression by decidual cells in preeclampsia: a potential blood biomarker.
- Author
-
Siddiqui MF, Nandi P, Girish GV, Nygard K, Eastabrook G, de Vrijer B, Han VK, and Lala PK
- Subjects
- Adult, Biomarkers metabolism, Case-Control Studies, Decidua cytology, Decorin genetics, Female, Humans, In Situ Hybridization, Polymerase Chain Reaction, Pregnancy, Pregnancy Trimester, Second, RNA, Messenger metabolism, Decidua metabolism, Decorin metabolism, Placenta metabolism, Pre-Eclampsia metabolism
- Abstract
Background: Decorin, a leucine-rich proteoglycan that is produced by decidual cells, limits invasion and endovascular differentiation of extravillous trophoblast cells during early placentation by binding to multiple tyrosine kinase receptors, in particular, vascular endothelial growth factor receptor-2., Objective: Because many studies have reported an association between poor trophoblast invasion and endovascular differentiation with preeclampsia, the studies reported here tested (1) whether decorin over-expression in the chorionic villi and/or basal decidua is associated with preeclampsia and, if so, (2) whether this association results in a hypoinvasive placenta, and (3) whether elevated plasma decorin concentration in the second trimester is a predictive biomarker for preeclampsia., Study Design: Decorin messenger RNA expression was measured with quantitative polymerase chain reaction at the tissue level and with in situ hybridization at the cellular level using (35)S-labeled antisense complimentary RNA probe in placentas from healthy control subjects and subjects with preeclampsia (14 each, 23-40 weeks of gestation). Tissue sections of the same placentas were also immunostained for decorin protein. A decorin over-expressing human endometrial stromal cell line was tested for invasion-regulatory effects on an invasive first-trimester extravillous trophoblast cell line HTR-8/SVneo plated in cocultures that were separated by a semipermeable membrane. Furthermore, we conducted retrospective measurements of plasma decorin levels during the second trimester (15-18 weeks of gestation) in a cohort of 28 body mass index-matched pairs of control subjects and subjects with preeclampsia before the onset of clinical disease., Results: First, decorin messenger RNA expression at the cellular level measured with in situ hybridization exhibited profoundly higher expression levels in basal plate decidual cells within the placentas from preeclamptic subjects than those from control subjects at all gestational ages, whereas no difference between the 2 subject groups was noted in villus mesenchymal cells. Similarly decorin messenger RNA expression at the tissue level in chorionic villi (primarily resulting from fetally derived mesenchymal cells) did not differ significantly between control and preeclampsia placentas. These findings were validated with immunostaining for decorin protein. Second, knocking down decorin gene in a decorin over-expressing endometrial cell line (used as an in vitro surrogate of decorin over-expressing decidual cells) in cocultures with extravillous trophoblast cells abrogated its invasion-restraining actions on trophoblast cells, which indicated paracrine contribution of decorin over-expressing decidua to the poor trophoblast invasiveness in situ. Finally, retrospective measurement of plasma decorin levels during the second trimester in 28 body mass index-matched pairs of control subjects and subjects with preeclampsia revealed elevated plasma decorin levels in all subjects with preeclampsia in all body mass index groups. A receiver operating characteristic curve analysis revealed strong diagnostic performance of plasma decorin in the prediction of preeclampsia status. Although there was no significant gestational age-related change in decorin levels during the second trimester in control or subjects with preeclampsia, we found that plasma decorin had a significant inverse relationship with body mass index or bodyweight., Conclusion: We conclude that decorin over-expression by basal decidual cells is associated with hypoinvasive phenotype and poor endovascular differentiation of trophoblast cells in preeclampsia and that elevated plasma decorin concentration is a potential predictive biomarker for preeclampsia before the onset of clinical signs., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
26. Mechanisms of trophoblast migration, endometrial angiogenesis in preeclampsia: The role of decorin.
- Author
-
Lala PK and Nandi P
- Subjects
- Animals, Endometrium pathology, Female, Humans, Pregnancy, Cell Movement, Decorin metabolism, Endometrium blood supply, Neovascularization, Physiologic, Pre-Eclampsia metabolism, Pre-Eclampsia pathology, Trophoblasts pathology
- Abstract
The objective of the present review is to synthesize the information on the cellular and molecular players responsible for maintaining a homeostatic balance between a naturally invasive human placenta and the maternal uterus in pregnancy; to review the roles of decorin (DCN) as a molecular player in this homeostasis; to list the common maladies associated with a break-down in this homeostasis, resulting from a hypo-invasive or hyper-invasive placenta, and their underlying mechanisms. We show that both the fetal components of the placenta, represented primarily by the extravillous trophoblast, and the maternal component represented primarily by the decidual tissue and the endometrial arterioles, participate actively in this balance. We discuss the process of uterine angiogenesis in the context of uterine arterial changes during normal pregnancy and preeclampsia. We compare and contrast trophoblast growth and invasion with the processes involved in tumorigenesis with special emphasis on the roles of DCN and raise important questions that remain to be addressed. Decorin (DCN) is a small leucine-rich proteoglycan produced by stromal cells, including dermal fibroblasts, chondrocytes, chorionic villus mesenchymal cells and decidual cells of the pregnant endometrium. It contains a 40 kDa protein core having 10 leucine-rich repeats covalently linked with a glycosaminoglycan chain. Biological functions of DCN include: collagen assembly, myogenesis, tissue repair and regulation of cell adhesion and migration by binding to ECM molecules or antagonising multiple tyrosine kinase receptors (TKR) including EGFR, IGF-IR, HGFR and VEGFR-2. DCN restrains angiogenesis by binding to thrombospondin-1, TGFβ, VEGFR-2 and possibly IGF-IR. DCN can halt tumor growth by antagonising oncogenic TKRs and restraining angiogenesis. DCN actions at the fetal-maternal interface include restraint of trophoblast migration, invasion and uterine angiogenesis. We demonstrate that DCN overexpression in the decidua is associated with preeclampsia (PE); this may have a causal role in PE by compromising endovascular differentiation of the trophoblast and uterine angiogenesis, resulting in poor arterial remodeling. Elevated DCN level in the maternal blood is suggested as a potential biomarker in PE.
- Published
- 2016
- Full Text
- View/download PDF
27. Restraint of Trophoblast Invasion of the Uterus by Decorin: Role in Pre-eclampsia.
- Author
-
Nandi P, Siddiqui MF, and Lala PK
- Subjects
- Animals, Biomarkers blood, Decidua metabolism, Decidua pathology, Decorin blood, Extracellular Signal-Regulated MAP Kinases immunology, Extracellular Signal-Regulated MAP Kinases metabolism, Female, Humans, Pre-Eclampsia blood, Pregnancy, Receptor Protein-Tyrosine Kinases immunology, Receptor Protein-Tyrosine Kinases metabolism, Trophoblasts metabolism, Trophoblasts pathology, Decidua immunology, Decorin immunology, Embryo Implantation immunology, MAP Kinase Signaling System immunology, Pre-Eclampsia immunology, Trophoblasts immunology
- Abstract
Decorin (DCN) is a leucine-rich, TGF-β binding proteoglycan produced by mesenchymal cells including chondrocytes, dermal fibroblasts, and uterine decidual cells. It exerts multiple physiological functions including collagen fibrillogenesis, myogenesis, angiostasis, and restraining placental invasiveness. We discovered that decidua-derived DCN restrains proliferation, migration, and invasion of extravillous trophoblast (EVT) cells of the human placenta in a TGF-β-independent manner. These functions were differentially mediated by binding of DCN to multiple tyrosine kinase receptors (TKR) including EGFR, IGFR1, and VEGFR2. DCN blocked VEGFR-2 dependent EVT cell migration and endovascular differentiation by inhibiting P38MAPK and ERK1/2 pathways.We identified the avid VEGFR2 binding site in DCN protein as a 12 amino acids (LGTNPLKSSGIE) span in the Leucine-rich-repeat (LRR) 5 region of domain III. A single amino acid mutation (substitution of K to A) of DCN at this site abrogated VEGFR-2- dependent DCN actions. Also, DCN mRNA expression, measured with in situ hybridization, was selectively upregulated in decidual cells in placentas from mothers suffering from pre-eclampsia (PE), whereas the expression levels remained unchanged in chorionic villus mesenchymal cells. This difference between PE and control placentas was present at all gestational ages, indicating the pathogenic role of DCN in PE. We hypothesize that increased blood DCN levels could be a candidate biomarker for PE., (© 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
28. COX-2 Elevates Oncogenic miR-526b in Breast Cancer by EP4 Activation.
- Author
-
Majumder M, Landman E, Liu L, Hess D, and Lala PK
- Subjects
- Animals, Cell Line, Tumor, Cell Movement, Female, Gene Expression drug effects, Humans, Lung Neoplasms secondary, Mice, Mice, Knockout, NF-kappa B metabolism, Neoplasm Invasiveness, Phosphatidylinositol 3-Kinases metabolism, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-akt metabolism, Receptors, Prostaglandin E metabolism, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cyclooxygenase 2 metabolism, MicroRNAs genetics, MicroRNAs metabolism, Receptors, Prostaglandin E, EP4 Subtype metabolism
- Abstract
Unlabelled: MicroRNAs (miRs) are small regulatory molecules emerging as potential biomarkers in cancer. Previously, it was shown that COX-2 expression promotes breast cancer progression via multiple mechanisms, including induction of stem-like cells (SLC), owing to activation of the prostaglandin E2 receptor EP4 (PTGER4). COX-2 overexpression also upregulated microRNA-526b (miR-526b), in association with aggressive phenotype. Here, the functional roles of miR-526b in breast cancer and the mechanistic role of EP4 signaling in miR-526b upregulation were examined. A positive correlation was noted between miR-526b and COX-2 mRNA expression in COX-2 disparate breast cancer cell lines. Stable overexpression of miR-526b in poorly metastatic MCF7 and SKBR3 cell lines resulted in increased cellular migration, invasion, EMT phenotype and enhanced tumorsphere formation in vitro, and lung colony formation in vivo in immunodeficient mice. Conversely, knockdown of miR-526b in aggressive MCF7-COX-2 and SKBR3-COX-2 cells reduced oncogenic functions and reversed the EMT phenotype, in vitro. Furthermore, it was determined that miR-526b expression is dependent on EP4 receptor activity and downstream PI3K-AKT and cyclic AMP (cAMP) signaling pathways. PI3K-AKT inhibitors blocked EP4 agonist-mediated miR-526b upregulation and tumorsphere formation in MCF7 and SKBR3 cells. NF-κB inhibitor abrogates EP agonist-stimulated miRNA expression in MCF7 and T47D cells, indicating that the NF-κB pathway is also involved in miR-526b regulation. In addition, inhibition of COX-2, EP4, PI3K, and PKA in COX-2-overexpressing cells downregulated miR-526b and its functions in vitro. Finally, miR-526b expression was significantly higher in cancerous than in noncancerous breast tissues and associated with reduced patient survival. In conclusion, miR-526b promotes breast cancer progression, SLC-phenotype through EP4-mediated signaling, and correlates with breast cancer patient survival., Implications: This study presents novel findings that miRNA 526b is a COX-2 upregulated, oncogenic miRNA promoting SLCs, the expression of which follows EP4 receptor-mediated signaling, and is a promising biomarker for monitoring and personalizing breast cancer therapy., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
29. The role of CCL21/CCR7 chemokine axis in breast cancer-induced lymphangiogenesis.
- Author
-
Tutunea-Fatan E, Majumder M, Xin X, and Lala PK
- Subjects
- Breast Neoplasms genetics, Cell Line, Tumor, Female, Gene Expression Regulation, Neoplastic, Humans, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction, Vascular Endothelial Growth Factor C genetics, Vascular Endothelial Growth Factor C metabolism, Breast Neoplasms metabolism, Breast Neoplasms pathology, Chemokine CCL21 metabolism, Lymphangiogenesis, Receptors, CCR7 metabolism
- Abstract
Background: Tumor-induced lymphangiogenesis facilitates breast cancer progression by generating new lymphatic vessels that serve as conduits for tumor dissemination to lymph nodes and beyond. Given the recent evidence suggesting the implication of C-C chemokine ligand 21/chemokine receptor 7 (CCL21/CCR7) in lymph node metastasis, the aim of our study was to define the role of this chemokine pair in breast cancer-associated lymphangiogenesis., Methods: The expression analysis of CCL21/CCR7 pair and lymphatic endothelial cell (LEC) markers in breast cancer specimens was performed by means of quantitative real-time PCR. By utilizing CCR7 and CCL21 gene manipulated breast cancer cell implants into orthotopic sites of nude mice, lymphatic vessel formation was assessed through quantitative real-time PCR, immunohistochemistry and immunofluorescence assays. Finally, the lymphangiogenic potential of CCL21/CCR7 was assessed in vitro with primary LECs through separate functional assays, each attempting to mimic different stages of the lymphangiogenic process., Results: We found that CCR7 mRNA expression in human breast cancer tissues positively correlates with the expression of lymphatic endothelial markers LYVE-1, podoplanin, Prox-1, and vascular endothelial growth factor-C (VEGF-C). We demonstrated that the expression of CCL21/CCR7 by breast cancer cells has the ability to promote tumor-induced lymph-vascular recruitment in vivo. In vitro, CCL21/CCR7 chemokine axis regulates the expression and secretion of lymphangiogenic factor VEGF-C and thereby promotes proliferation, migration, as well as tube formation of the primary human LECs. Finally, we showed that protein kinase B (AKT) signaling pathway is the intracellular mechanism of CCR7-mediated VEGF-C secretion by human breast cancer cells., Conclusions: These results reveal that CCR7 and VEGF-C display a significant crosstalk and suggest a novel role of the CCL21/CCR7 chemokine axis in the promotion of breast cancer-induced lymphangiogenesis.
- Published
- 2015
- Full Text
- View/download PDF
30. Prostaglandin E2 receptor EP4 as the common target on cancer cells and macrophages to abolish angiogenesis, lymphangiogenesis, metastasis, and stem-like cell functions.
- Author
-
Majumder M, Xin X, Liu L, Girish GV, and Lala PK
- Subjects
- Adenocarcinoma blood supply, Adenocarcinoma secondary, Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Apoptosis, Benzamides pharmacology, Benzamides therapeutic use, Celecoxib, Cell Line, Tumor, Cell Proliferation, Cyclooxygenase 2 metabolism, Drug Screening Assays, Antitumor, Female, Lung Neoplasms blood supply, Lung Neoplasms secondary, Lymphangiogenesis, Lymphatic Metastasis, Mammary Neoplasms, Experimental blood supply, Mammary Neoplasms, Experimental pathology, Mice, Mice, Inbred C3H, Molecular Targeted Therapy, Neoplasm Transplantation, Neoplastic Stem Cells, Pyrazoles pharmacology, Pyrazoles therapeutic use, Receptors, Prostaglandin E, EP4 Subtype metabolism, Sulfonamides pharmacology, Sulfonamides therapeutic use, Tumor Burden, Vascular Endothelial Growth Factor A metabolism, Adenocarcinoma drug therapy, Lung Neoplasms drug therapy, Macrophages metabolism, Mammary Neoplasms, Experimental drug therapy, Neovascularization, Pathologic drug therapy, Receptors, Prostaglandin E, EP4 Subtype antagonists & inhibitors
- Abstract
We previously established that COX-2 overexpression promotes breast cancer progression and metastasis. As long-term use of COX-2 inhibitors (COX-2i) can promote thrombo-embolic events, we tested an alternative target, prostaglandin E2 receptor EP4 subtype (EP4), downstream of COX-2. Here we used the highly metastatic syngeneic murine C3L5 breast cancer model to test the role of EP4-expressing macrophages in vascular endothelial growth factor (VEGF)-C/D production, angiogenesis, and lymphangiogenesis in situ, the role of EP4 in stem-like cell (SLC) functions of tumor cells, and therapeutic effects of an EP4 antagonist RQ-15986 (EP4A). C3L5 cells expressed all EP receptors, produced VEGF-C/D, and showed high clonogenic tumorsphere forming ability in vitro, functions inhibited with COX-2i or EP4A. Treating murine macrophage RAW 264.7 cell line with COX-2i celecoxib and EP4A significantly reduced VEGF-A/C/D production in vitro, measured with quantitative PCR and Western blots. Orthotopic implants of C3L5 cells in C3H/HeJ mice showed rapid tumor growth, angiogenesis, lymphangiogenesis (CD31/LYVE-1 and CD31/PROX1 immunostaining), and metastasis to lymph nodes and lungs. Tumors revealed high incidence of EP4-expressing, VEGF-C/D producing macrophages identified with dual immunostaining of F4/80 and EP4 or VEGF-C/D. Celecoxib or EP4A therapy at non-toxic doses abrogated tumor growth, lymphangiogenesis, and metastasis to lymph nodes and lungs. Residual tumors in treated mice revealed markedly reduced VEGF-A/C/D and phosphorylated Akt/ERK proteins, VEGF-C/D positive macrophage infiltration, and proliferative/apoptotic cell ratios. Knocking down COX-2 or EP4 in C3L5 cells or treating cells in vitro with celecoxib or EP4A and treating tumor-bearing mice in vivo with the same drug reduced SLC properties of tumor cells including preferential co-expression of COX-2 and SLC markers ALDH1A, CD44, OCT-3/4, β-catenin, and SOX-2. Thus, EP4 is an excellent therapeutic target to block stem-like properties, angiogenesis, and lymphangiogenesis induced by VEGF-A/C/D secreted by cancer cells and tumor infiltrating macrophages., (© 2014 The Authors. Cancer Science published by Wiley Publishing Asia Pty Ltd on behalf of Japanese Cancer Association.)
- Published
- 2014
- Full Text
- View/download PDF
31. Pharmacogenetics of chronic pain management.
- Author
-
Kapur BM, Lala PK, and Shaw JL
- Subjects
- Drug Interactions genetics, Humans, Pain Management methods, Pharmacogenetics methods, Adjuvants, Pharmaceutic therapeutic use, Analgesics, Opioid therapeutic use, Chronic Pain drug therapy, Chronic Pain genetics
- Abstract
Objective: The experience of chronic pain is one of the commonest reasons individuals seek medical attention, making the management of chronic pain a major issue in clinical practice. Drug metabolism and responses are affected by many factors, with genetic variations offering only a partial explanation of an individual's response. There is a paucity of evidence for the benefits of pharmacogenetic testing in the context of pain management., Design and Methods: We reviewed the literature between 2000 and 2013, and references cited therein, using various keywords related to pain management, pharmacology and pharmacogenetics., Results: Opioids continue to be the mainstay of chronic pain management. Several non-opioid based therapies, such as treatment with cannabinoids, gene therapy and epigenetic-based approaches are now available for these patients. Adjuvant therapies with antidepressants, benzodiazepines or anticonvulsants can also be useful in managing pain. Currently, laboratory monitoring of pain management patients, if performed, is largely through urine drug measurements., Conclusions: Drug half-life calculations can be used as functional markers of the cumulative effect of pharmacogenetics and drug-drug interactions. Assessment of half-life and therapeutic effects may be more useful than genetic testing in preventing adverse drug reactions to pain medications, while ensuring effective analgesia. Definitive, mass spectrometry-based methods, capable of measuring parent drug and metabolite levels, are the most useful assays for this purpose. Urine drug measurements do not necessarily correlate with serum drug concentrations or therapeutic effects. Therefore, they are limited in their use in monitoring efficacy and toxicity., (Copyright © 2014 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
32. A practical and sensitive method of quantitating lymphangiogenesis in vivo.
- Author
-
Majumder M, Xin X, and Lala PK
- Subjects
- Angiogenesis Inhibitors analysis, Animals, Breast Neoplasms blood supply, Celecoxib, Cell Line, Tumor, Cyclooxygenase 2 Inhibitors, Female, Gene Knockdown Techniques, Glycoproteins metabolism, Humans, Membrane Transport Proteins, Mice, Mice, Inbred C3H, Mice, Nude, Neovascularization, Pathologic, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Pyrazoles, RNA, Messenger metabolism, Sulfonamides, Vascular Endothelial Growth Factor D genetics, Breast Neoplasms metabolism, Lymphangiogenesis, Molecular Imaging methods, Vascular Endothelial Growth Factor D metabolism
- Abstract
To address the inadequacy of current assays, we developed a directed in vivo lymphangiogenesis assay (DIVLA) by modifying an established directed in vivo angiogenesis assay. Silicon tubes (angioreactors) were implanted in the dorsal flanks of nude mice. Tubes contained either growth factor-reduced basement membrane extract (BME)-alone (negative control) or BME-containing vascular endothelial growth factor (VEGF)-D (positive control for lymphangiogenesis) or FGF-2/VEGF-A (positive control for angiogenesis) or a high VEGF-D-expressing breast cancer cell line MDA-MD-468LN (468-LN), or VEGF-D-silenced 468LN. Lymphangiogenesis was detected superficially with Evans Blue dye tracing and measured in the cellular contents of angioreactors by multiple approaches: lymphatic vessel endothelial hyaluronan receptor-1 (Lyve1) protein (immunofluorescence) and mRNA (qPCR) expression and a visual scoring of lymphatic vs blood capillaries with dual Lyve1 (or PROX-11 or Podoplanin)/Cd31 immunostaining in cryosections. Lymphangiogenesis was absent with BME, high with VEGF-D or VEGF-D-producing 468LN cells and low with VEGF-D-silenced 468LN. Angiogenesis was absent with BME, high with FGF-2/VEGF-A, moderate with 468LN or VEGF-D and low with VEGF-D-silenced 468LN. The method was reproduced in a syngeneic murine C3L5 tumor model in C3H/HeJ mice with dual Lyve1/Cd31 immunostaining. Thus, DIVLA presents a practical and sensitive assay of lymphangiogenesis, validated with multiple approaches and markers. It is highly suited to identifying pro- and anti-lymphangiogenic agents, as well as shared or distinct mechanisms regulating lymphangiogenesis vs angiogenesis, and is widely applicable to research in vascular/tumor biology.
- Published
- 2013
- Full Text
- View/download PDF
33. Mechanisms in decorin regulation of vascular endothelial growth factor-induced human trophoblast migration and acquisition of endothelial phenotype.
- Author
-
Lala N, Girish GV, Cloutier-Bosworth A, and Lala PK
- Subjects
- Cells, Cultured, Drug Antagonism, Endothelium, Vascular cytology, Endothelium, Vascular physiology, Female, Human Umbilical Vein Endothelial Cells cytology, Human Umbilical Vein Endothelial Cells drug effects, Human Umbilical Vein Endothelial Cells physiology, Humans, Neovascularization, Physiologic drug effects, Neovascularization, Physiologic physiology, Phenotype, Pregnancy, Signal Transduction drug effects, Trophoblasts physiology, Vascular Endothelial Growth Factor A antagonists & inhibitors, Cell Differentiation drug effects, Cell Movement drug effects, Decorin pharmacology, Endothelium, Vascular drug effects, Trophoblasts drug effects, Vascular Endothelial Growth Factor A pharmacology
- Abstract
Extravillous trophoblast (EVT) cells of the human placenta invade the uterine decidua and utero-placental arteries to establish an efficient exchange of key molecules between maternal and fetal blood. Trophoblast invasion is stringently regulated in situ both positively and negatively by a variety of factors at the fetal-maternal interface to maintain a healthy utero-placental homeostasis. One such factor, decorin, a transforming growth factor (TGF)-beta binding, leucine-rich proteoglycan produced by the decidua, negatively regulates EVT proliferation, migration, and invasiveness independent of TGF-beta. We reported that these decorin actions were mediated by its binding to multiple tyrosine kinase receptors, including vascular endothelial growth factor receptor (VEGFR)-2. The present study explores the mechanisms underlying decorin antagonism of VEGF (VEGF-A) stimulation of endovascular differentiation of EVT using our EVT cell line, HTR-8/SVneo. We observe that decorin inhibits VEGF-induced EVT cell migration and endothelial-like tube formation on matrigel. VEGF activates MAPKs (p38 MAPK, MEK3/6, and ERK1/2) in EVT cells, and the activation is blocked in both cases by decorin. Employing selective MAPK inhibitors, we show that both p38 and ERK pathways contribute independently to VEGF-induced EVT migration and capillary-like tube formation. VEGF upregulates the vascular endothelial (VE) markers VE-cadherin and beta-catenin in EVT and endothelial cells, and this upregulation is blocked by decorin and MAPK inhibitors. These results suggest that decorin inhibits VEGF-A stimulation of trophoblast migration and endovascular differentiation by interfering with p38 MAPK and ERK1/2 activation. Thus decorin-mediated dual impediment of endovascular differentiation of the EVT and angiogenesis may have implications for pathogenesis of preeclampsia, a hypoinvasive trophoblast disorder in pregnancy.
- Published
- 2012
- Full Text
- View/download PDF
34. An in vitro evaluation of antimicrobial efficacy of primary root canal filling materials.
- Author
-
Hegde S, Lala PK, Dinesh RB, and Shubha AB
- Subjects
- Bacillus subtilis drug effects, Bacterial Load drug effects, Barium Sulfate pharmacology, Calcium Hydroxide pharmacology, Candida albicans drug effects, Colony Count, Microbial, Drug Combinations, Enterococcus faecalis drug effects, Escherichia coli drug effects, Humans, Hydrocarbons, Iodinated pharmacology, Materials Testing, Pseudomonas aeruginosa drug effects, Silicone Oils pharmacology, Sodium Fluoride pharmacology, Staphylococcus aureus drug effects, Staphylococcus epidermidis drug effects, Streptococcus mutans drug effects, Zinc Oxide pharmacology, Zinc Oxide-Eugenol Cement pharmacology, Anti-Infective Agents pharmacology, Root Canal Filling Materials pharmacology
- Abstract
Objective: The present study aimed to evaluate and compare six different materials commonly used for filling the root canals of primary teeth for antimicrobial efficacy against some of the microorganisms commonly found in infected root canals., Study Design: In this experimental in vitro study six root canal filling materials were tested for antimicrobial efficacy against eight microbial strains using the agar diffusion method., Results: Zinc oxide eugenol paste exhibited the strongest antimicrobial potential followed by Endoflas, zinc oxide-calcium hydroxide-sodium fluoride mixture, zinc oxide-calcium hydroxide mixture and calcium hydroxide paste (Apexcal). The addition of sodium fluoride to the zinc oxide-calcium hydroxide mixture enhanced the antimicrobial efficacy. Metapex demonstrated minimal inhibition and Vaseline was non-inhibitory., Conclusions: All the test filling materials demonstrated varying antimicrobial activity against the microorganisms tested. Zinc oxide eugenol paste and materials containing zinc oxide were found to be more effective against the microorganisms compared to materials without zinc oxide.
- Published
- 2012
- Full Text
- View/download PDF
35. Targeting COX-2 and EP4 to control tumor growth, angiogenesis, lymphangiogenesis and metastasis to the lungs and lymph nodes in a breast cancer model.
- Author
-
Xin X, Majumder M, Girish GV, Mohindra V, Maruyama T, and Lala PK
- Subjects
- Animals, Apoptosis drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Female, Lymphangiogenesis drug effects, Mammary Neoplasms, Experimental blood supply, Mammary Neoplasms, Experimental metabolism, Mice, Mice, Inbred C3H, Molecular Targeted Therapy, Neoplasm Metastasis, Neovascularization, Pathologic drug therapy, Proto-Oncogene Proteins c-akt metabolism, Vascular Endothelial Growth Factor C metabolism, Vascular Endothelial Growth Factor D metabolism, Cyclooxygenase 2 metabolism, Cyclooxygenase 2 Inhibitors pharmacology, Lung Neoplasms secondary, Mammary Neoplasms, Experimental drug therapy, Mammary Neoplasms, Experimental pathology, Receptors, Prostaglandin E, EP4 Subtype antagonists & inhibitors
- Abstract
We reported that cyclo-oxygenase (COX)-2 expression in human breast cancer stimulated cancer cell migration and invasiveness, production of vascular endothelial growth factor (VEGF)-C and lymphangiogenesis in situ, largely from endogenous PGE2-mediated stimulation of prostaglandin E (EP)1 and EP4 receptors, presenting them as candidate therapeutic targets against lymphatic metastasis. As human breast cancer xenografts in immuno-compromised mice have limitations for preclinical testing, we developed a syngeneic murine breast cancer model of spontaneous lymphatic metastasis mimicking human and applied it for mechanistic and therapeutic studies. We tested the roles of COX-2 and EP receptors in VEGF-C and -D production by a highly metastatic COX-2 expressing murine breast cancer cell line C3L5. These cells expressed all EP receptors and produced VEGF-C and -D, both inhibited with COX-2 inhibitors or EP4 (but not EP1, EP2 or EP3) antagonists. C3H/HeJ mice, when implanted SC in both inguinal regions with C3L5 cells suspended in growth factor-reduced Matrigel, exhibited rapid tumor growth, tumor-associated angiogenesis and lymphangiogenesis (respectively measured with CD31 and LYVE-1 immunostaining), metastasis to the inguinal and axillary lymph nodes and the lungs. Chronic oral administration of COX-1/COX-2 inhibitor indomethacin, COX-2 inhibitor celecoxib and an EP4 antagonist ONO-AE3-208, but not an EP1 antagonist ONO-8713 at nontoxic doses markedly reduced tumor growth, lymphangiogenesis, angiogenesis, and metastasis to lymph nodes and lungs. Residual tumors in responding mice revealed reduced VEGF-C and -D proteins, AkT phosphorylation and increased apoptotic/proliferative cell ratios consistent with blockade of EP4 signaling. We suggest that EP4 antagonists deserve clinical testing for chemo-intervention of lymphatic metastasis in human breast cancer.
- Published
- 2012
- Full Text
- View/download PDF
36. Co-expression of α9β1 integrin and VEGF-D confers lymphatic metastatic ability to a human breast cancer cell line MDA-MB-468LN.
- Author
-
Majumder M, Tutunea-Fatan E, Xin X, Rodriguez-Torres M, Torres-Garcia J, Wiebe R, Timoshenko AV, Bhattacharjee RN, Chambers AF, and Lala PK
- Subjects
- Animals, Breast cytology, Breast metabolism, Breast pathology, Cell Line, Tumor, Cell Movement, Cell Proliferation, Female, Humans, Lymphatic Metastasis, Mice, Mice, Nude, Neoplasm Invasiveness genetics, Breast Neoplasms genetics, Breast Neoplasms pathology, Gene Expression Regulation, Neoplastic, Integrins genetics, Vascular Endothelial Growth Factor D genetics
- Abstract
Introduction and Objectives: Lymphatic metastasis is a common occurrence in human breast cancer, mechanisms remaining poorly understood. MDA-MB-468LN (468LN), a variant of the MDA-MB-468GFP (468GFP) human breast cancer cell line, produces extensive lymphatic metastasis in nude mice. 468LN cells differentially express α9β1 integrin, a receptor for lymphangiogenic factors VEGF-C/-D. We explored whether (1) differential production of VEGF-C/-D by 468LN cells provides an autocrine stimulus for cellular motility by interacting with α9β1 and a paracrine stimulus for lymphangiogenesis in vitro as measured with capillary-like tube formation by human lymphatic endothelial cells (HMVEC-dLy); (2) differential expression of α9 also promotes cellular motility/invasiveness by interacting with macrophage derived factors; (3) stable knock-down of VEGF-D or α9 in 468LN cells abrogates lymphangiogenesis and lymphatic metastasis in vivo in nude mice., Results: A comparison of expression of cyclo-oxygenase (COX)-2 (a VEGF-C/-D inducer), VEGF-C/-D and their receptors revealed little COX-2 expression by either cells. However, 468LN cells showed differential VEGF-D and α9β1 expression, VEGF-D secretion, proliferative, migratory/invasive capacities, latter functions being stimulated further with VEGF-D. The requirement of α9β1 for native and VEGF-D-stimulated proliferation, migration and Erk activation was demonstrated by treating with α9β1 blocking antibody or knock-down of α9. An autocrine role of VEGF-D in migration was shown by its impairment by silencing VEGF-D and restoration with VEGF-D. 468LN cells and their soluble products stimulated tube formation, migration/invasiveness of HMVEC-dLy cell in a VEGF-D dependent manner as indicated by the loss of stimulation by silencing VEGF-D in 468LN cells. Furthermore, 468LN cells showed α9-dependent stimulation of migration/invasiveness by macrophage products. Finally, capacity for intra-tumoral lymphangiogenesis and lymphatic metastasis in nude mice was completely abrogated by stable knock-down of either VEGF-D or α9 in 468LN cells., Conclusion: Differential capacity for VEGF-D production and α9β1 integrin expression by 468LN cells jointly contributed to their lymphatic metastatic phenotype.
- Published
- 2012
- Full Text
- View/download PDF
37. Decorin is a novel VEGFR-2-binding antagonist for the human extravillous trophoblast.
- Author
-
Khan GA, Girish GV, Lala N, Di Guglielmo GM, and Lala PK
- Subjects
- Amino Acid Sequence, Animals, Blotting, Far-Western, Cattle, Cell Line, Cell Movement drug effects, Cell Proliferation drug effects, Decorin chemistry, Enzyme Activation drug effects, Extracellular Signal-Regulated MAP Kinases metabolism, HLA-G Antigens metabolism, Humans, Immunoprecipitation, Ligands, Models, Biological, Molecular Sequence Data, Peptides chemistry, Peptides metabolism, Protein Binding drug effects, Protein Structure, Tertiary, Trophoblasts cytology, Trophoblasts drug effects, Trophoblasts enzymology, Vascular Endothelial Growth Factor A pharmacology, Vascular Endothelial Growth Factor Receptor-2 chemistry, Vascular Endothelial Growth Factor Receptor-2 metabolism, Decorin metabolism, Trophoblasts metabolism, Vascular Endothelial Growth Factor Receptor-2 antagonists & inhibitors
- Abstract
Extravillous trophoblasts (EVT) of the human placenta invade the uterine decidua and its arteries to ensure successful placentation. We previously identified two decidua-derived molecules, TGF-β and a TGF-β-binding proteoglycan decorin (DCN), as negative regulators of EVT proliferation, migration, and invasiveness and reported that DCN acts via multiple tyrosine kinase receptors [epidermal growth factor-receptor (EGF-R), IGF receptor-1 (IGFR1), and vascular endothelial growth factor 2 receptor (VEGFR-2)]. Because binding of DCN to VEGFR-2 has never been reported earlier, present study explored this binding, the approximate location of VEGFR-2-binding site in DCN, and its functional role in our human first trimester EVT cell line HTR-8/SVneo. Based on far-Western blotting and coimmunoprecipitation studies, we report that DCN binds both native (EVT expressed) and recombinant VEGFR-2 and that this binding is abrogated with a VEGFR-2 blocking antibody, indicating an overlap between the ligand-binding and the DCN-binding domains of VEGFR-2. We determined that (125)I-labeled VEGF-E (a VEGFR-2 specific ligand) binds EVT with a dissociation constant (K(d)) of 566 pM, and DCN displaced this binding with an inhibition constant (K(i)) of 3.93-5.78 nM, indicating a 7- to 10-fold lower affinity of DCN for VEGFR-2. DCN peptide fragments derived from the leucine rich repeat 5 domain that blocked DCN-VEGFR-2 interactions or VEGF-E binding in EVT cells also blocked VEGF-A- and VEGF-E-induced EVT cell proliferation and migration, indicative of functional VEGFR-2-binding sites of DCN. Finally, DCN inhibited VEGF-E-induced EVT migration by interfering with ERK1/2 activation. Our findings reveal a novel role of DCN as an antagonistic ligand for VEGFR-2, having implications for pathophysiology of preeclampsia, a trophoblast hypoinvasive disorder in pregnancy, and explain its antiangiogenic function.
- Published
- 2011
- Full Text
- View/download PDF
38. IFPA Meeting 2010 Workshops Report II: Placental pathology; trophoblast invasion; fetal sex; parasites and the placenta; decidua and embryonic or fetal loss; trophoblast differentiation and syncytialisation.
- Author
-
Al-Khan A, Aye IL, Barsoum I, Borbely A, Cebral E, Cerchi G, Clifton VL, Collins S, Cotechini T, Davey A, Flores-Martin J, Fournier T, Franchi AM, Fretes RE, Graham CH, Godbole G, Hansson SR, Headley PL, Ibarra C, Jawerbaum A, Kemmerling U, Kudo Y, Lala PK, Lassance L, Lewis RM, Menkhorst E, Morris C, Nobuzane T, Ramos G, Rote N, Saffery R, Salafia C, Sarr D, Schneider H, Sibley C, Singh AT, Sivasubramaniyam TS, Soares MJ, Vaughan O, Zamudio S, and Lash GE
- Subjects
- Animals, Cell Differentiation physiology, Cell Fusion, Cell Movement physiology, Decidua physiology, Decidua physiopathology, Education, Female, Humans, Male, Parasitic Diseases immunology, Parasitic Diseases metabolism, Parasitic Diseases pathology, Parasitic Diseases physiopathology, Placenta Accreta etiology, Placenta Accreta metabolism, Placenta Accreta pathology, Placenta Accreta physiopathology, Pregnancy, Pregnancy Complications metabolism, Pregnancy Complications physiopathology, Pregnancy Outcome, Sex Characteristics, Stress, Physiological physiology, Trophoblasts cytology, Fetus cytology, Fetus parasitology, Fetus pathology, Fetus physiology, Fetus physiopathology, Placenta cytology, Placenta parasitology, Placenta pathology, Placenta physiology, Placenta physiopathology, Trophoblasts physiology
- Abstract
Workshops are an important part of the IFPA annual meeting. At IFPA Meeting 2010 diverse topics were discussed in twelve themed workshops, six of which are summarized in this report. 1. The placental pathology workshop focused on clinical correlates of placenta accreta/percreta. 2. Mechanisms of regulation of trophoblast invasion and spiral artery remodeling were discussed in the trophoblast invasion workshop. 3. The fetal sex and intrauterine stress workshop explored recent work on placental sex differences and discussed them in the context of whether boys live dangerously in the womb.4. The workshop on parasites addressed inflammatory responses as a sign of interaction between placental tissue and parasites. 5. The decidua and embryonic/fetal loss workshop focused on key regulatory mediators in the decidua, embryo and fetus and how alterations in expression may contribute to different diseases and adverse conditions of pregnancy. 6. The trophoblast differentiation and syncytialisation workshop addressed the regulation of villous cytotrophoblast differentiation and how variations may lead to placental dysfunction and pregnancy complications., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
39. Differential stimulation of VEGF-C production by adhesion/growth-regulatory galectins and plant lectins in human breast cancer cells.
- Author
-
Timoshenko AV, Kaltner H, André S, Gabius HJ, and Lala PK
- Subjects
- Cell Line, Tumor, Culture Media, Serum-Free, Female, Humans, Neovascularization, Pathologic metabolism, Breast Neoplasms blood supply, Breast Neoplasms metabolism, Galectins pharmacology, Plant Lectins pharmacology, Vascular Endothelial Growth Factor C biosynthesis
- Abstract
Unlabelled: The present study tested the hypothesis that the production of vascular endothelial growth factor C (VEGF-C), a key lymphangiogenic factor, by human breast cancer cells can be stimulated by human lectins, using plant lectins as controls., Materials and Methods: The effects of human galectins and five plant lectins reacting with distinct determinants of N- and O-glycans on the accumulation of VEGF-C in serum-free cell culture media of human breast cancer cells endowed with high (MDA-MB-231) and low (MCF7, T-47D, and SK-BR-3) VEGF-C-producing abilities were examined., Results: All tested lectins stimulated VEGF-C production by MDA-MB-231 cells, albeit with different potency. Concanavalin A, but not galectins, was also able to stimulate VEGF-C production by low VEGF-C-producing cell lines MCF7 and T-47D. Both VEGF-C mRNA and protein were strongly up-regulated in SK-BR-3 cells by concanavalin A and wheat germ agglutinin, but not jacalin., Conclusion: The differential response of breast cancer cell lines separated by the endogenous level of VEGF-C production suggests that galectins may contribute to tumor-associated lymphangiogenesis in a cell-specific manner.
- Published
- 2010
40. Relationship between cyclooxygenase-2 and human epidermal growth factor receptor 2 in vascular endothelial growth factor C up-regulation and lymphangiogenesis in human breast cancer.
- Author
-
Bhattacharjee RN, Timoshenko AV, Cai J, and Lala PK
- Subjects
- Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Line, Tumor, Cyclooxygenase 2 genetics, Female, Humans, Immunohistochemistry, RNA Interference, RNA, Messenger genetics, RNA, Messenger metabolism, Receptor, ErbB-2 genetics, Reverse Transcriptase Polymerase Chain Reaction, Up-Regulation, Vascular Endothelial Growth Factor C genetics, Vesicular Transport Proteins genetics, Vesicular Transport Proteins metabolism, Breast Neoplasms metabolism, Cyclooxygenase 2 metabolism, Lymphangiogenesis, Receptor, ErbB-2 metabolism, Vascular Endothelial Growth Factor C metabolism
- Abstract
Both cyclooxygenase (COX)-2 and human epidermal growth factor receptor (HER)-2 promote breast cancer progression; however, the relationship between the two molecules remains unclear. We utilized human breast cancer tissues and cell lines to examine whether COX-2 and HER-2 played independent or interdependent roles in vascular endothelial growth factor (VEGF)-C up-regulation and lymphangiogenesis. A paired correlation of immunodetectable levels of COX-2, VEGF-C, and HER-2 proteins and lymphovascular density (LVD; D2-40-immunolabeled) in 55 breast cancer specimens revealed a positive correlation between COX-2 and HER-2 irrespective of clinicopathological status. However COX-2 alone positively correlated with LVD. In 10 independent specimens, mRNA levels showed a positive correlation between HER-2 and COX-2 or VEGF-C but not LYVE-1 (lymphovascular endothelial marker). These findings implicate COX-2, but not HER-2, in breast cancer-associated lymphangiogenesis. Manipulation of the COX-2 or HER-2 genes in breast cancer cell lines varying widely in COX-2 and HER-2 expression revealed a direct role of COX-2 and an indirect COX-2 dependent role of HER-2 in VEGF-C up-regulation: (i) high VEGF-C expression in high COX-2/low HER-2 expressing MDA-MB-231 cells was reduced by siRNA-mediated down-regulation of COX-2, but not HER-2; (ii) integration of HER-2 in these cells simultaneously up-regulated COX-2 protein as well as VEGF-C secretion; and (iii) low VEGF-C secretion by high HER-2/low COX-2 expressing SK-BR-3 cells was stimulated by COX-2 overexpression. These findings of the primary role of COX-2 and the COX-2-dependent role of HER-2, if any, in VEGF-C up-regulation and lymphangiogenesis suggest that COX-2 inhibitors may abrogate lymphatic metastasis in breast cancer irrespective of HER-2 status., (© 2010 Japanese Cancer Association.)
- Published
- 2010
- Full Text
- View/download PDF
41. Decorin-mediated inhibition of proliferation and migration of the human trophoblast via different tyrosine kinase receptors.
- Author
-
Iacob D, Cai J, Tsonis M, Babwah A, Chakraborty C, Bhattacharjee RN, and Lala PK
- Subjects
- Cell Line, Cell Movement physiology, Chorionic Villi metabolism, Decorin, ErbB Receptors antagonists & inhibitors, ErbB Receptors physiology, Female, Fibronectins pharmacology, Humans, Immunoblotting, Phosphorylation drug effects, Pregnancy, Receptor Protein-Tyrosine Kinases antagonists & inhibitors, Receptor, IGF Type 1 antagonists & inhibitors, Receptor, IGF Type 1 physiology, Transforming Growth Factor beta immunology, Transforming Growth Factor beta metabolism, Trophoblasts cytology, Trophoblasts metabolism, Vascular Endothelial Growth Factor Receptor-2 antagonists & inhibitors, Vascular Endothelial Growth Factor Receptor-2 physiology, Cell Movement drug effects, Cell Proliferation drug effects, Extracellular Matrix Proteins pharmacology, Proteoglycans pharmacology, Receptor Protein-Tyrosine Kinases physiology, Trophoblasts drug effects
- Abstract
Decorin (DCN), a decidua-derived TGFbeta-binding proteoglycan, negatively regulates proliferation, migration, and invasiveness of human extravillous trophoblast (EVT) cells in a TGFbeta-independent manner. The present study examined underlying mechanisms, in particular possible roles of epidermal growth factor receptor (EGFR), IGF receptor (IGFR)-I, and vascular endothelial growth factor receptor (VEGFR)-2. EVT cell sprouting from first-trimester chorionic villus explants in the presence or absence of TGFbeta-neutralizing antibody was inhibited with DCN, suggesting its negative regulatory role in situ. Inhibition of migration of the human EVT cell line HTR-8/SVneo in transwells undercoated with fibronectin was stronger when cells were briefly preincubated with DCN at 4 C (known to retard dissociation of receptor-ligand complex) than at 37 C, suggesting possible DCN action by cell membrane binding. Pretreatment of cells with an IGFR-I blocking agent, but not two EGFR blocking agents or a VEGFR blocking agent, significantly abrogated migration inhibitory effects of DCN, suggesting the involvement of IGFR-I but not EGFR or VEGFR in migration inhibition by DCN. On the other hand, pretreatment with either of the EGFR blocking agents, or the VEGFR blocking agent but not the IGFR-I blocking agent, blocked proliferation inhibitory effects of DCN, indicating the roles of EGFR and VEGFR, but not IGFR-I in antiproliferative action of DCN. EVT cells expressed EGFR, IGFR-I, and VEGFR-2. IGFR-I and VEGF-R2 were phosphorylated in the presence of their natural ligands as well as DCN, and these events were blocked by pretreatment with respective receptor blocking agents indicating DCN-mediated activation of these receptors. In conclusion, DCN effects on EVT cells are mediated selectively by multiple tyrosine kinase receptors.
- Published
- 2008
- Full Text
- View/download PDF
42. Prostaglandin E2-mediated migration of human trophoblast requires RAC1 and CDC42.
- Author
-
Nicola C, Lala PK, and Chakraborty C
- Subjects
- Aminoquinolines pharmacology, Base Sequence, Cell Line, Cell Movement drug effects, Cell Movement physiology, Dinoprostone pharmacology, Enzyme Inhibitors pharmacology, Female, Humans, Pregnancy, Pyrimidines pharmacology, RNA, Small Interfering genetics, Tissue Culture Techniques, Trophoblasts drug effects, cdc42 GTP-Binding Protein antagonists & inhibitors, cdc42 GTP-Binding Protein genetics, rac1 GTP-Binding Protein antagonists & inhibitors, rac1 GTP-Binding Protein genetics, Dinoprostone physiology, Trophoblasts cytology, Trophoblasts physiology, cdc42 GTP-Binding Protein physiology, rac1 GTP-Binding Protein physiology
- Abstract
The invasion of maternal decidua and uterine spiral arteries by a trophoblast subpopulation called extravillous trophoblast (EVT) is essential for the establishment of a normal placenta and an adequate blood flow toward the fetus. Derangements in these processes underlie pregnancy-related diseases like preeclampsia and intrauterine growth restriction. Many growth factors, growth factor binding proteins, and extracellular matrix components can positively or negatively regulate the proliferation, migration, and/or invasiveness of these EVT cells. RHO GTPases, including RHOA, RAC1, and CDC42, are ubiquitous proteins that control cytoskeletal changes by forming stress fibers and projecting lamellipodia and filopodia during cellular migration. We had previously shown that prostaglandin (PG) E(2) produced in abundance by the decidua promotes the migration of first-trimester human EVTs by increasing the intracellular concentration of calcium and activating calpain. Using our well-characterized immortalized EVT cell line, HTR-8/SVneo, as well as villus explants from first-trimester placentae, this study examined the role of RHO GTPases RAC1 and CDC42 in PGE(2)-mediated migratory responses of these cells. Though a RAC1 inhibitor, NSC23766 as well as RAC1 knockdown by siRNA decreased the migration of HTR-8/SVneo cells in a Transwell migration assay, this inhibition could not be restored by PGE(2) or 17-phenyl trinor PGE(2) (PGE receptor PTGER1 agonist) or PGE(1) Alcohol (PGE receptor PTGER4 agonist). Similar results were noted for EVT cell spreading in villus explants. Furthermore, CDC42 silencing using siRNA inhibited PGE(2)-induced migration of HTR-8/SVneo cells. Finally, the treatment of EVT cells with PGE(2), PTGER1 agonist, or PTGER4 agonist activated RAC1 and CDC42 at 10 min, suggesting that RAC1 and CDC42 play an essential role in PGE(2)-mediated migration of human EVTs.
- Published
- 2008
- Full Text
- View/download PDF
43. Roles of Rho guanosine 5'-triphosphatase A, Rho kinases, and extracellular signal regulated kinase (1/2) in prostaglandin E2-mediated migration of first-trimester human extravillous trophoblast.
- Author
-
Nicola C, Chirpac A, Lala PK, and Chakraborty C
- Subjects
- Amides pharmacology, Butadienes pharmacology, Cell Line, Cell Movement drug effects, Chorionic Villi metabolism, Chorionic Villi pathology, Enzyme Inhibitors pharmacology, Female, Humans, Mitogen-Activated Protein Kinase 1 antagonists & inhibitors, Mitogen-Activated Protein Kinase 1 drug effects, Mitogen-Activated Protein Kinase 3 antagonists & inhibitors, Mitogen-Activated Protein Kinase 3 drug effects, Nitriles pharmacology, Pregnancy, Pyridines pharmacology, RNA, Small Interfering pharmacology, Trophoblasts pathology, rho-Associated Kinases antagonists & inhibitors, rho-Associated Kinases drug effects, rhoA GTP-Binding Protein antagonists & inhibitors, rhoA GTP-Binding Protein drug effects, Dinoprostone metabolism, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 metabolism, Pregnancy Trimester, First metabolism, Trophoblasts metabolism, rho-Associated Kinases metabolism, rhoA GTP-Binding Protein metabolism
- Abstract
Prostaglandin (PG) E(2) may regulate invasiveness of human placenta because we previously reported stimulation of migration of placental trophoblasts by PGE(2) acting through PGE receptor (EP)-1 and activating calpain. RhoA GTPase and its important effector Rho kinase (ROCK) have also been previously shown to regulate trophoblast migration. Using immortalized HTR-8/SVneo trophoblast cells and first-trimester human chorionic villus explant cultures on matrigel, we further examined the role of RhoA/ROCK and MAPK (ERK1/2) pathways on PGE(2)-mediated stimulation of trophoblast migration. Migration of cytotrophoblasts was shown to be inhibited by treatment of the trophoblast cell line and chorionic villus explants with either cell-permeable C3 transferase or selective RhoA small interfering RNA. These inhibitions were significantly mitigated by the addition of PGE(2), an EP1/EP3 agonist or an EP3/EP4 agonist, suggesting that RhoA plays an important role in trophoblast migration but may not be obligatory for PGE(2) action. Treatment of HTR-8/SVneo cells with nonselective ROCK inhibitor Y27632 or ROCK small interfering RNAs inhibited migration of these cells, which could not be rescued with PGE(2) or the other two EP agonists, suggesting the obligatory role of ROCK in PGE(2)-induced migratory response. Furthermore, U0126, an inhibitor of MAPK kinases MEK1 and MEK2, abrogated PGE(2)-induced migration of trophoblasts, and PGE(2) or the other two EP agonists stimulated ERK1/2 activation in trophoblasts, which was not abrogated by pretreatment with C3 transferase, indicating that ERK signaling pathway is an efficient alternate pathway for RhoA in PGE(2)-mediated migration of trophoblasts. These results suggest that ROCK and ERK1/2 play more important roles than RhoA in PGE(2)-mediated migration stimulation of first-trimester trophoblasts.
- Published
- 2008
- Full Text
- View/download PDF
44. Migration-promoting role of VEGF-C and VEGF-C binding receptors in human breast cancer cells.
- Author
-
Timoshenko AV, Rastogi S, and Lala PK
- Subjects
- Cell Line, Tumor, Cell Movement, Female, Flow Cytometry, Humans, Neuropilin-1 metabolism, Neuropilin-2 metabolism, RNA, Small Interfering, Reverse Transcriptase Polymerase Chain Reaction, Transfection, Breast Neoplasms metabolism, Breast Neoplasms pathology, Neoplasm Metastasis pathology, Receptors, Vascular Endothelial Growth Factor metabolism, Vascular Endothelial Growth Factor C metabolism
- Abstract
Vascular endothelial growth factor C (VEGF-C) is a lymphangiogenic factor over-expressed in highly metastatic, cyclooxygenase (COX)-2 expressing breast cancer cells. We tested the hypothesis that tumour-derived VEGF-C may play an autocrine role in metastasis by promoting cellular motility through one or more VEGF-C-binding receptors VEGFR-2, VEGFR-3, neuropilin (NRP)-1, NRP-2, and integrin alpha9beta1. We investigated the expression of these receptors in several breast cancer cell lines (MDA-MB-231, Hs578T, SK-BR-3, T-47D, and MCF7) and their possible requirement in migration of two VEGF-C-secreting, highly metastatic lines MDA-MB-231 and Hs578T. While cell lines varied significantly in their expression of above VEGF-C receptors, migratory activity of MDA-MB-231 and Hs578T cells was linked to one or more of these receptors. Depletion of endogenous VEGF-C by treatments with a neutralising antibody, VEGF-C siRNA or inhibitors of Src, EGFR/Her2/neu and p38 MAP kinases which inhibited VEGF-C production, inhibited cellular migration, indicating the requirement of VEGF-C for migratory function. Migration was differentially attenuated by blocking or downregulation of different VEGF-C receptors, for example treatment with a VEGFR-2 tyrosine kinase inhibitor, NRP-1 and NRP-2 siRNA or alpha9beta1 integrin antibody, indicating the participation of one or more of the receptors in cell motility. This novel role of tumour-derived VEGF-C indicates that breast cancer metastasis can be promoted by coordinated stimulation of lymphangiogenesis and enhanced migratory activity of breast cancer cells.
- Published
- 2007
- Full Text
- View/download PDF
45. Effects of PGE2 on human trophoblast proliferation and migration.
- Author
-
Lala PK, Nicola C, and Chakraborty C
- Subjects
- Cell Line, Female, Humans, Cell Movement drug effects, Cell Proliferation drug effects, Dinoprostone pharmacology, Trophoblasts drug effects
- Published
- 2006
- Full Text
- View/download PDF
46. COX-2-mediated stimulation of the lymphangiogenic factor VEGF-C in human breast cancer.
- Author
-
Timoshenko AV, Chakraborty C, Wagner GF, and Lala PK
- Subjects
- Cell Line, Tumor, Cyclooxygenase 2 biosynthesis, Cyclooxygenase 2 drug effects, Cyclooxygenase Inhibitors pharmacology, Down-Regulation, Enzyme Activation drug effects, Enzyme Activation physiology, Female, Glycoproteins biosynthesis, Humans, Imidazoles pharmacology, Molecular Sequence Data, Pyrazoles pharmacology, Pyridines pharmacology, Pyrimidines pharmacology, Quinazolines pharmacology, RNA, Messenger biosynthesis, RNA, Messenger metabolism, RNA, Small Interfering pharmacology, Receptors, Prostaglandin E metabolism, Receptors, Prostaglandin E physiology, Receptors, Prostaglandin E, EP1 Subtype, Receptors, Prostaglandin E, EP4 Subtype, Structure-Activity Relationship, Vascular Endothelial Growth Factor C antagonists & inhibitors, Vascular Endothelial Growth Factor C biosynthesis, Vesicular Transport Proteins, Breast Neoplasms metabolism, Cyclooxygenase 2 physiology, Lymphangiogenesis physiology, Vascular Endothelial Growth Factor C metabolism
- Abstract
Increased expression of COX-2 or VEGF-C has been correlated with progressive disease in certain cancers. Present study utilized several human breast cancer cell lines (MCF-7, T-47D, Hs578T and MDA-MB-231, varying in COX-2 expression) as well as 10 human breast cancer specimens to examine the roles of COX-2 and prostaglandin E (EP) receptors in VEGF-C expression or secretion, and the relationship of COX-2 or VEGF-C expression to lymphangiogenesis. We found a strong correlation between COX-2 mRNA expression and VEGF-C expression or secretion levels in breast cancer cell lines and VEGF-C expression in breast cancer tissues. Expression of LYVE-1, a selective marker for lymphatic endothelium, was also positively correlated with COX-2 or VEGF-C expression in breast cancer tissues. Inhibition of VEGF-C expression and secretion in the presence of COX-1/2 or COX-2 inhibitors or following downregulation of COX-2 with COX-2 siRNA established a stimulatory role COX-2 in VEGF-C synthesis by breast cancer cells. EP1 as well as EP4 receptor antagonists inhibited VEGF-C production indicating the roles of EP1 and EP4 in VEGF-C upregulation by endogenous PGE2. Finally, VEGF-C secretion by MDA-MB-231 cells was inhibited in the presence of kinase inhibitors for Her-2/neu, Src and p38 MAPK, indicating a requirement of these kinases for VEGF-C synthesis. These results, for the first time, demonstrate a regulatory role of COX-2 in VEGF-C synthesis (and thereby lymphangiogenesis) in human breast cancer, which is mediated at least in part by EP1/EP4 receptors.
- Published
- 2006
- Full Text
- View/download PDF
47. EP1 receptor-mediated migration of the first trimester human extravillous trophoblast: the role of intracellular calcium and calpain.
- Author
-
Nicola C, Timoshenko AV, Dixon SJ, Lala PK, and Chakraborty C
- Subjects
- Calcium Signaling drug effects, Calcium Signaling physiology, Cell Movement drug effects, Cells, Cultured, Chelating Agents pharmacology, Chorionic Villi metabolism, Dinoprostone metabolism, Dinoprostone pharmacology, Egtazic Acid analogs & derivatives, Egtazic Acid pharmacology, Enzyme Activation drug effects, Enzyme Inhibitors pharmacology, Female, Humans, Phenotype, Pregnancy, Pregnancy Trimester, First, Receptors, Prostaglandin E genetics, Receptors, Prostaglandin E, EP1 Subtype, Reverse Transcriptase Polymerase Chain Reaction, Thapsigargin pharmacology, Type C Phospholipases antagonists & inhibitors, Calcium metabolism, Calpain metabolism, Cell Movement physiology, Receptors, Prostaglandin E metabolism, Trophoblasts cytology, Trophoblasts physiology
- Abstract
Context: The root cause of preeclampsia in the human lies in the placenta, where a subpopulation of cytotrophoblast cells called extravillous trophoblasts (EVT), known to be involved in the invasion of the uterine endometrium and utero-placental arteries, become less invasive, resulting in poor perfusion of maternal blood into placenta., Objectives: Because EVT migrate into the prostaglandin (PG) E2-rich decidua, we tested the roles of PGE2 and PGE2-mediated signaling in EVT migration, using our well-characterized EVT line HTR-8/Svneo as well as first trimester villus explants in culture., Design: mRNA expression of different PGE2 receptors (EPs) in HTR-8/Svneo cells was studied using RT-PCR. To characterize the functional significance of EP receptors in EVT, different EP receptor agonists and antagonists were used in our migration assay systems and in the measurements of intracellular concentration of Ca2+ ([Ca2+]i) and calpain activity., Results: Exogenous PGE2 stimulated EVT migration both in vitro and in the villus explant cultures. Although EVT expressed mRNA for all EP receptors (EP 1-4), a functional predominance of EP1 and EP4 was demonstrated in migration assays using specific EP agonists and antagonists. EP1-receptor-mediated signaling events such as activation of phospholipase C and elevation of cytosolic free [Ca2+]i were confirmed by the following findings: 1) exogenous PGE2 or an EP1 agonist, but not an EP4 agonist, increased [Ca2+]i, which could be blocked with an EP1 antagonist as well as BAPTA and thapsigargin; 2) phospholipase C inhibitor U73122, BAPTA, and thapsigargin inhibited PGE2-mediated migratory response of EVT; and 3) PGE2-mediated EVT migration was shown to be dependent on a class of Ca2+-dependent proteases called calpains, known to be involved in cell detachment from substratum during migratory responses. The presence of PGE2 stimulated calpain activity, whereas two calpain inhibitors, calpastatin and N-Ac-Leu-Leu-methioninal (ALLM), blocked EVT migration., Conclusion: PGE2 stimulates EVT migration by signaling through EP1 receptors, increasing [Ca2+]i, and activating calpain.
- Published
- 2005
- Full Text
- View/download PDF
48. HCG increases trophoblast migration in vitro via the insulin-like growth factor-II/mannose-6 phosphate receptor.
- Author
-
Zygmunt M, McKinnon T, Herr F, Lala PK, and Han VK
- Subjects
- Antibodies, Blocking pharmacology, Antibodies, Monoclonal pharmacology, Cells, Cultured, Chorionic Gonadotropin pharmacology, Humans, Insulin-Like Growth Factor I genetics, Insulin-Like Growth Factor I immunology, Insulin-Like Growth Factor I metabolism, Insulin-Like Growth Factor II genetics, Insulin-Like Growth Factor II immunology, Placenta cytology, Placenta drug effects, Placenta physiology, RNA, Messenger analysis, RNA, Messenger metabolism, Receptor, IGF Type 2 immunology, Cell Movement drug effects, Chorionic Gonadotropin physiology, Insulin-Like Growth Factor II metabolism, Receptor, IGF Type 2 metabolism, Trophoblasts physiology
- Abstract
We have previously shown that both HCG and insulin-like growth factor-II (IGF-II) stimulate trophoblastic invasion. Furthermore, the invasion-promoting function of IGF-II resulted from IGF-II mannose 6-phosphate receptor (IGF-II/M6PR) activation. Since HCG and IGF-II did not have an additive effect on cell migration of extravillous trophoblast (EVT) cell line, HTR-8 SVneo, we hypothesized that HCG actions are mediated via alterations in the expression and/or function of IGF-II axis. HCG treatment (50-50,000 mU/ml) of the HTR-8/SVneo cells did not alter the expression of either insulin-like growth factor-I or IGF-II mRNA or peptide synthesis, but caused (i) an increase in the (125)I-IGF-II binding to EVT cells, and (ii) an increase in the externalization rate of the IGF-II binding sites without affecting their internalization. This effect was due to the increase in the number of IGF-II binding sites in the plasma membrane without any change in the IGF-II binding affinity. Although HCG did not influence the abundance of IGF-II/M6PR mRNA or protein, anti-IGF-II/M6PR antibody decreased HCG-induced migration of EVT, supporting the hypothesis that HCG might stimulate EVT migration by increasing IGF-II binding to the plasma membrane and subsequently by increasing the IGF-II effect probably mediated via the IGF-II/M6PR.
- Published
- 2005
- Full Text
- View/download PDF
49. Nodal and ALK7 inhibit proliferation and induce apoptosis in human trophoblast cells.
- Author
-
Munir S, Xu G, Wu Y, Yang B, Lala PK, and Peng C
- Subjects
- Activin Receptors, Type I genetics, Apoptosis physiology, Base Sequence, Cell Division physiology, Cell Line, DNA Primers genetics, DNA-Binding Proteins genetics, DNA-Binding Proteins physiology, Female, G1 Phase, Gene Expression, Humans, Nodal Protein, Pregnancy, RNA, Messenger genetics, RNA, Messenger metabolism, Signal Transduction, Smad2 Protein, Smad3 Protein, Trans-Activators genetics, Trans-Activators physiology, Transfection, Transforming Growth Factor beta genetics, Trophoblasts physiology, Activin Receptors, Type I physiology, Transforming Growth Factor beta physiology, Trophoblasts cytology
- Abstract
Nodal, a member of the transforming growth factor-beta superfamily, is known to play critical roles in early vertebrate development, but its functions in extraembryonic tissues are unclear. ALK7 is a type I receptor for Nodal. Recently, we demonstrated that Nodal mRNA and several ALK7 transcripts are expressed in human placenta throughout pregnancy (Roberts, H. J., Hu, S., Qiu, Q., Leung, P. C. K., Cannigia, I., Gruslin, A., Tsang, B., and Peng, C. (2003) Biol. Reprod. 68, 1719-1726). In this study, we determined the role of Nodal and ALK7 in trophoblast cell proliferation and apoptosis. Overexpression of Nodal in normal trophoblast cells (HTR8/SVneo) and several choriocarcinoma cell lines resulted in a significant decrease in the number of metabolically active cells. The effect of Nodal could be mimicked by constitutively active ALK7 (ALK7-ca), but was blocked by kinase-deficient ALK7. The growth inhibitory effect of Nodal was also blocked by dominant-negative Smad2/3. Overexpression of Nodal and ALK7-ca induced apoptosis in trophoblast cells as determined by Hoechst staining, flow cytometry, and caspase-3 Western blotting. In addition, Nodal and ALK7-ca decreased the number of proliferating cells as measured by bromodeoxyuridine assays. Furthermore, overexpression of Nodal or ALK7-ca increased p27 expression, but reduced the levels of Cdk2 and cyclin D(1). Taken together, this study demonstrates for the first time that Nodal, acting through ALK7 and Smad2/3, inhibits proliferation and induces apoptosis in human trophoblast cells. Our findings also suggest that the Nodal-ALK7 pathway inhibits cell proliferation by inducing G(1) cell cycle arrest and that this effect is mediated in part by the p27-cyclin E/Cdk2 pathway.
- Published
- 2004
- Full Text
- View/download PDF
50. PGE2-mediated upregulation of iNOS in murine breast cancer cells through the activation of EP4 receptors.
- Author
-
Timoshenko AV, Lala PK, and Chakraborty C
- Subjects
- Animals, Cyclic AMP pharmacology, Cyclooxygenase 2, Cyclooxygenase 2 Inhibitors, Cyclooxygenase Inhibitors pharmacology, Enzyme Induction, Enzyme Inhibitors pharmacology, Female, Gene Expression Regulation, Enzymologic, Humans, Interferon-gamma pharmacology, Isoenzymes metabolism, Lipopolysaccharides pharmacology, Mammary Neoplasms, Experimental pathology, Membrane Proteins, Mice, Mice, Inbred C3H, Nitric Oxide metabolism, Nitric Oxide Synthase antagonists & inhibitors, Nitric Oxide Synthase genetics, Nitric Oxide Synthase Type II, Prostaglandin-Endoperoxide Synthases metabolism, Prostaglandins E metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Receptors, Prostaglandin E agonists, Receptors, Prostaglandin E antagonists & inhibitors, Receptors, Prostaglandin E, EP4 Subtype, Tumor Cells, Cultured, Up-Regulation, Dinoprostone physiology, Mammary Neoplasms, Experimental enzymology, Nitric Oxide Synthase metabolism, Receptors, Prostaglandin E metabolism
- Abstract
We report here that endogenous prostaglandin E(2) (PGE(2)) resulting from cyclooxygenase (COX)-2 expression in a highly metastatic murine breast cancer cell line C3L5 upregulates IFN-gamma + LPS-induced nitric oxide (NO) synthase (iNOS) expression and NO production. This action of PGE(2) is mediated through the EP(4) receptor in a cAMP-dependent manner. Both nonselective and selective COX-2 inhibitors suppressed IFN-gamma + LPS-induced NO production, which was largely restored by exogenous PGE(2) or EP(4) receptor agonist PGE(1) alcohol. EP(4) antagonist AH-23848B inhibited NO production with a concomitant downregulation of iNOS mRNA in IFN-gamma + LPS-stimulated cells. cAMP dependence of NO production by cells under inducible conditions was demonstrated by the use of known modulators of intracellular cAMP. Since both COX-2 and iNOS are implicated in breast cancer progression, our findings of EP(4) receptor-mediated upregulation of iNOS in COX-2-expressing breast cancer cells suggest that blocking COX-2 and/or EP(4) may provide a simple therapeutic modality in this tumor model., (Copyright 2003 Wiley-Liss, Inc.)
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.