1
|
La Padula S, Pensato R, Errico C, D'Andrea F, Persichetti P, Chesné C, Jamin A, Ben Mosbah I, Toia F, Cordova A, Meningaud JP, Hersant B. Effects of Nanofat and PRP on Type I Collagen Production in Striae Distensae: Preliminary Findings from a Prospective, Randomized Single-Blind Study. Aesthetic Plast Surg 2025; 49:2076-2084. [PMID: 39627329 DOI: 10.1007/s00266-024-04560-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 11/14/2024] [Indexed: 04/26/2025]
Abstract
INTRODUCTION Striae distensae (SD) appear clinically as parallel striae, lying perpendicular to the tension lines of the skin. SD evolve into two clinical phases, an initial inflammatory phase in which they are called "striae rubrae" (SR) and a chronic phase in which they are called striae albae (SA). This study investigates the synergistic effect of nanofat and platelet-rich plasma (PRP) injections on collagen production in fibroblasts derived from SA (SAF). MATERIAL AND METHODS A prospective, randomized single-blind study was conducted in fifty women presenting with SA in the abdominal region who had voluntarily sought a conventional abdominoplasty procedure and accepted to test an autologous treatment for their SDs. SA were treated using: PrP 10 ml; PrP 2ml (20%) + nanofat 8ml (80%); nanofat 10ml. Following the abdominal dermolipectomy, biopsies from treated and untreated SDs were taken and analyzed for type I collagen quantification. Results were processed through statistical analysis models using the Student's t test. RESULTS Collagen concentration in untreated SA biopsies was significantly lower than in healthy skin. Both PRP and nanofat treatments significantly increased collagen biosynthesis compared to controls, with the combined PRP-nanofat treatment showing the highest increase in collagen levels (p < 0.0001). A superior clinical improvement was observed in the areas that received the combined treatment of PRP and nanofat (p = 0.001). CONCLUSION Our findings indicate that both PRP and nanofat treatments effectively enhance collagen production in SA, with the combined PRP-nanofat treatment showing a synergistic effect. This combined therapy holds promise for effectively treating SA, providing a new potential treatment avenue for SMs and similar skin conditions. Further studies are needed to validate these results and explore clinical applications. LEVEL OF EVIDENCE I This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Simone La Padula
- Department of Plastic and Reconstructive Surgery, Università degli studi di Napoli Federico II, Via Pansini 5, 80131, Napoli, Italy.
- Department of Plastic, Reconstructive and Maxillo facial Surgery, Henri Mondor Hospital, University Paris XII, 51 Avenue du Maréchal de Lattre de Tassigny, 94000, Créteil, France.
- , 50 rue Saint, 75011 PARIS, Sébastien, France.
| | - Rosita Pensato
- Department of Plastic and Reconstructive Surgery, Università degli studi di Napoli Federico II, Via Pansini 5, 80131, Napoli, Italy
| | - Concetta Errico
- Department of Plastic and Reconstructive Surgery, Università degli studi di Napoli Federico II, Via Pansini 5, 80131, Napoli, Italy
| | - Francesco D'Andrea
- Department of Plastic and Reconstructive Surgery, Università degli studi di Napoli Federico II, Via Pansini 5, 80131, Napoli, Italy
| | - Paolo Persichetti
- Department of Plastic and Reconstructive Surgery, Università Campus Biomedico di Roma, Via Alvaro del Portillo, 21, 00128, Roma, Italy
| | - Christophe Chesné
- Biopredic International, Parc d'activité de la Bretèche Bâtiment A4, 35760, Saint Grégoire, France
| | - Agnes Jamin
- Biopredic International, Parc d'activité de la Bretèche Bâtiment A4, 35760, Saint Grégoire, France
| | - Ismail Ben Mosbah
- Biopredic International, Parc d'activité de la Bretèche Bâtiment A4, 35760, Saint Grégoire, France
- IMRB - Institut Mondor de Recherche Biomédicale. 51 Avenue du Maréchal de Lattre de Tassigny, 94000, Créteil, France
| | - Francesca Toia
- Plastic and Reconstructive Surgery, Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Adriana Cordova
- Plastic and Reconstructive Surgery, Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Jean Paul Meningaud
- Department of Plastic, Reconstructive and Maxillo facial Surgery, Henri Mondor Hospital, University Paris XII, 51 Avenue du Maréchal de Lattre de Tassigny, 94000, Créteil, France
| | - Barbara Hersant
- Department of Plastic, Reconstructive and Maxillo facial Surgery, Henri Mondor Hospital, University Paris XII, 51 Avenue du Maréchal de Lattre de Tassigny, 94000, Créteil, France
| |
Collapse
|
2
|
Freer M, Cooper J, Goncalves K, Przyborski S. Bioengineering the Human Intestinal Mucosa and the Importance of Stromal Support for Pharmacological Evaluation In Vitro. Cells 2024; 13:1859. [PMID: 39594608 PMCID: PMC11592477 DOI: 10.3390/cells13221859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Drug discovery is associated with high levels of compound elimination in all stages of development. The current practices for the pharmacokinetic testing of intestinal absorption combine Transwell® inserts with the Caco-2 cell line and are associated with a wide range of limitations. The improvement of pharmacokinetic research relies on the development of more advanced in vitro intestinal constructs that better represent human native tissue and its response to drugs, providing greater predictive accuracy. Here, we present a humanized, bioengineered intestinal construct that recapitulates aspects of intestinal microanatomy. We present improved histotypic characteristics reminiscent of the human intestine, such as a reduction in transepithelial electrical resistance (TEER) and the formation of a robust basement membrane, which are contributed to in-part by a strong stromal foundation. We explore the link between stromal-epithelial crosstalk, paracrine communication, and the role of the keratinocyte growth factor (KGF) as a soluble mediator, underpinning the tissue-specific role of fibroblast subpopulations. Permeability studies adapted to a 96-well format allow for high throughput screening and demonstrate the role of the stromal compartment and tissue architecture on permeability and functionality, which is thought to be one of many factors responsible for unexpected drug outcomes using current approaches for pharmacokinetic testing.
Collapse
Affiliation(s)
- Matthew Freer
- Department of Biosciences, Durham University, Durham DH1 3LE, UK;
| | - Jim Cooper
- European Collection of Authenticated Cell Cultures, Salisbury SP4 0JG, UK;
| | - Kirsty Goncalves
- Department of Biosciences, Durham University, Durham DH1 3LE, UK;
| | - Stefan Przyborski
- Department of Biosciences, Durham University, Durham DH1 3LE, UK;
- Reprocell Europe Ltd., Glasgow G20 0XA, UK
| |
Collapse
|
3
|
Jerman UD, Veranič P, Cirman T, Kreft ME. Human Amniotic Membrane Enriched with Urinary Bladder Fibroblasts Promote the Re-Epithelization of Urothelial Injury. Cell Transplant 2021; 29:963689720946668. [PMID: 32841052 PMCID: PMC7563929 DOI: 10.1177/0963689720946668] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Culturing cells in three-dimensional systems that include extracellular matrix
components and different cell types mimic the native tissue and as such provide
much more representative results than conventional two-dimensional cell
cultures. In order to develop biomimetic bladder tissue in vitro, we used human
amniotic membrane (AM) extracellular matrix as a scaffold for bladder
fibroblasts (BFs) and urothelial cells. Our aims were to evaluate the
integration of BFs into the AM stroma, to assess the differentiation of the
urothelium on BFs-enriched AM scaffolds, and to evaluate the AM as a urothelial
wound dressing. First, to achieve the optimal integration of BFs into AM stroma,
different intact and de- epithelialized AM (dAM) scaffolds were tested. BFs
secreted matrix metalloproteinase (MMP)-1 and MMP-2 and integrated into the
stroma of all types of AM scaffolds. Second, to establish urothelial tissue
equivalent, urothelial cells were seeded on dAM scaffolds enriched with BFs. The
BFs in the stroma of the AM scaffolds promoted (1) the proliferation of
urothelial cells, (2) the attachment of urothelial cells on AM basal lamina with
hemidesmosomes, and (3) development of multilayered urothelium with expressed
uroplakins and well-developed cell junctions. Third, we established an ex vivo
model of the injured bladder to evaluate the dAM as a wound dressing for
urothelial full-thickness injury. dAM acted as a promising wound dressing since
it enabled rapid re-epithelization of urothelial injury and integrated into the
bladder tissue. Herein, the developed urothelial tissue equivalents enable
further mechanistic studies of bladder epithelial–mesenchymal interactions, and
they could be applied as biomimetic models for preclinical testing of newly
developed drugs. Moreover, we could hypothesize that AM may be suitable as a
dressing of the wound that occurs during transurethral resection of bladder
tumor, since it could diminish the possibility of tumor recurrence, by promoting
the rapid re-epithelization of the urothelium.
Collapse
Affiliation(s)
- Urška Dragin Jerman
- Institute of Cell Biology, 37664Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Peter Veranič
- Institute of Cell Biology, 37664Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tina Cirman
- 86684Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Mateja Erdani Kreft
- Institute of Cell Biology, 37664Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
4
|
La Padula S, Hersant B, Pizza C, Chesné C, Jamin A, Ben Mosbah I, Errico C, D'Andrea F, Rega U, Persichetti P, Meningaud JP. Striae Distensae: In Vitro Study and Assessment of Combined Treatment With Sodium Ascorbate and Platelet-Rich Plasma on Fibroblasts. Aesthetic Plast Surg 2021; 45:1282-1293. [PMID: 33474574 DOI: 10.1007/s00266-020-02100-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/13/2020] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Striae distensae (SD) appear clinically as parallel striae, lying perpendicular to the tension lines of the skin. SD evolve into two clinical phases, an initial inflammatory phase in which they are called "striae rubrae" (SR) and a chronic phase in which they are called striae albae (SA). Fibroblasts seem to play a key role in the pathogenesis of stretch marks. This study was aimed at describing and analyzing stretch marks-derived fibroblasts (SMF), the differences between SR- and SA-derived fibroblasts (SRF, SAF), testing two treatments in vitro (sodium ascorbate and PrP) on SAF. MATERIAL AND METHODS To characterize the SMF, the expression of alpha smooth muscle actin (alpha SMA) was investigated. Type I collagen expression was measured in SAF, before and after adding different PrP concentrations and sodium ascorbate in the culture medium. Results were processed through statistical analysis models using the Student's t-test. RESULTS A significant increase in alpha SMA (P <0.001) was observed in SRF. SAF treated with PrP and sodium ascorbate showed a resumption of their metabolic activity by an increase in collagen type I production and cell proliferation. After 24 h of incubation with PrP 1% and PrP 5% + sodium ascorbate, cell viability was increased by 140% and 151% and by 156 and 178% after 48 h, respectively, compared to the control. CONCLUSION Our study shows that a biologically mediated improvement in SMF metabolic activity is possible. Our promising results require further trials to be able to confirm the reproducibility of this combined treatment, particularly in vivo. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable.
Collapse
Affiliation(s)
- Simone La Padula
- Department of Plastic, Reconstructive and Maxillo facial Surgery, Henri Mondor Hospital, University Paris XII, 51 Avenue du Maréchal de Lattre de Tassigny, 94000, Créteil, France.
- Department of Plastic and Reconstructive Surgery, Università Campus Biomedico di Roma, Via Alvaro del Portillo, 21, 00128, Roma, Italy.
- Department of Plastic and Reconstructive Surgery, Università degli studi di Napoli Federico II, Via Pansini 5, 80131, Napoli, Italy.
- , 50 rue Saint Sébastien, Paris, 75011, France.
| | - Barbara Hersant
- Department of Plastic, Reconstructive and Maxillo facial Surgery, Henri Mondor Hospital, University Paris XII, 51 Avenue du Maréchal de Lattre de Tassigny, 94000, Créteil, France
| | - Chiara Pizza
- Department of Plastic, Reconstructive and Maxillo facial Surgery, Henri Mondor Hospital, University Paris XII, 51 Avenue du Maréchal de Lattre de Tassigny, 94000, Créteil, France
| | - Christophe Chesné
- Biopredic International, Parc d'activité de la Bretèche Bâtiment A4, 35760, Saint Grégoire, France
| | - Agnes Jamin
- Biopredic International, Parc d'activité de la Bretèche Bâtiment A4, 35760, Saint Grégoire, France
| | - Ismail Ben Mosbah
- Biopredic International, Parc d'activité de la Bretèche Bâtiment A4, 35760, Saint Grégoire, France
- IMRB - Institut Mondor de Recherche Biomédicale, 51 Avenue du Maréchal de Lattre de Tassigny, 94000, Créteil, France
| | - Concetta Errico
- Department of Plastic, Reconstructive and Maxillo facial Surgery, Henri Mondor Hospital, University Paris XII, 51 Avenue du Maréchal de Lattre de Tassigny, 94000, Créteil, France
| | - Francesco D'Andrea
- Department of Plastic and Reconstructive Surgery, Università degli studi di Napoli Federico II, Via Pansini 5, 80131, Napoli, Italy
| | - Umberto Rega
- Department of Plastic and Reconstructive Surgery, Università degli studi di Napoli Federico II, Via Pansini 5, 80131, Napoli, Italy
| | - Paolo Persichetti
- Department of Plastic and Reconstructive Surgery, Università Campus Biomedico di Roma, Via Alvaro del Portillo, 21, 00128, Roma, Italy
| | - Jean Paul Meningaud
- Department of Plastic, Reconstructive and Maxillo facial Surgery, Henri Mondor Hospital, University Paris XII, 51 Avenue du Maréchal de Lattre de Tassigny, 94000, Créteil, France
| |
Collapse
|
5
|
Liu F, Li Z, Guo J, Fang S, Zhou J, Cao B, Liu J, Yi Y, Yuan X, Xu X, Huang O, Wang L, Zou Y. Endometrial stromal cell proteomic analysis reveals LIM and SH3 protein 1 (LASP1) plays important roles in the progression of adenomyosis. Mol Hum Reprod 2021; 27:6129094. [PMID: 33543750 DOI: 10.1093/molehr/gaab008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 01/12/2021] [Indexed: 11/14/2022] Open
Abstract
Adenomyosis is one of the most common gynecological disorders that the molecular events underlying its pathogenesis remain not fully understood. Prior studies have shown that endometrial stromal cells (ESCs) played crucial roles in the pathogenesis of adenomyosis. In this study, we utilized two-dimensional gel electrophoresis combined with protein identification by mass spectrometry (2D/MS) proteomics analysis to compare the differential protein expression profile between the paired eutopic and ectopic ESCs (EuESCs and EcESCs) in adenomyosis, and a total of 32 significantly altered protein spots were identified. Among which, the expression of LIM and SH3 protein 1 (LASP1) was increased significantly in EcESCs compared to EuESCs. Immunohistochemical assay showed that LASP1 was overexpressed in the stromal cells of ectopic endometriums compared to eutopic endometriums; further functional analyses revealed that LASP1 overexpression could enhance cell proliferation, migration and invasion of EcESCs. Furthermore, we also showed that the dysregulated expression of LASP1 in EcESCs was associated with DNA hypermethylation in the promoter region of the LASP1 gene. However, the detailed molecular mechanisms of enhancing cell proliferation, invasion and migration caused by upregulated LASP1 in adenomyosis needs further study. For the first time, our data suggested that LASP1 plays important roles in the pathogenesis of adenomyosis, and could serve as a prognostic biomarker of adenomyosis.
Collapse
Affiliation(s)
- Faying Liu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Zengming Li
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Jiubai Guo
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Department of Gynecology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Shufen Fang
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Department of Gynecology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Jiangyan Zhou
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Department of Gynecology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Bianna Cao
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Department of Gynecology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Jun Liu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Department of Gynecology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Yulan Yi
- Department of Gynecology, Huangshi Central Hospital, Huangshi, Hubei, China
| | - Xiaoqun Yuan
- Department of Gynecology, Jiujiang Maternal and Child Health Hospital, Jiujiang, Jiangxi, China
| | - Xiaoyun Xu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Department of Gynecology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Ouping Huang
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Department of Gynecology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Liqun Wang
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Department of Reproductive Health, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Yang Zou
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| |
Collapse
|
6
|
Effect of naive and cancer-educated fibroblasts on colon cancer cell circadian growth rhythm. Cell Death Dis 2020; 11:289. [PMID: 32341349 PMCID: PMC7184765 DOI: 10.1038/s41419-020-2468-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/18/2020] [Accepted: 02/18/2020] [Indexed: 01/28/2023]
Abstract
Opportunistic modification of the tumour microenvironment by cancer cells enhances tumour expansion and consequently eliminates tumour suppressor components. We studied the effect of fibroblasts on the circadian rhythm of growth and protein expression in colon cancer HCT116 cells and found diminished oscillation in the proliferation of HCT116 cells co-cultured with naive fibroblasts, compared with those co-cultured with tumour-associated fibroblasts (TAFs) or those cultured alone, suggesting that TAFs may have lost or gained factors that regulate circadian phenotypes. Based on the fibroblast paracrine factor analysis, we tested IL6, which diminished HCT116 cell growth oscillation, inhibited early phase cell proliferation, increased early phase expression of the differentiation markers CEA and CDX2, and decreased early phase ERK5 phosphorylation. In conclusion, our data demonstrate how the cancer education of naive fibroblasts influences the circadian parameters of neighbouring cancer cells and highlights a putative role for IL6 as a novel candidate for preoperative treatments.
Collapse
|
7
|
Szczepańska AA, Łupicka M, Socha BM, Korzekwa AJ. The influence of arachidonic acid metabolites on PPAR and RXR expression in bovine uterine cells. Gen Comp Endocrinol 2018; 262:27-35. [PMID: 29510153 DOI: 10.1016/j.ygcen.2018.03.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 02/22/2018] [Accepted: 03/02/2018] [Indexed: 02/05/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors belonging to the superfamily of nuclear receptors. Three isoforms have been described: alpha (PPARα), delta (PPARδ), and gamma (PPARγ). PPARs heterodimerize with retinoid X receptors (RXRs: RXRα, RXRβ and RXRγ). PPAR activity can be modulated by several ligands, including arachidonic acid (AA) metabolites. The aims of the study were to determine the effect of AA metabolites (prostaglandin [PG]E2, PGF2α, leukotriene [LT]B4, and LTC4) on mRNA (real-time PCR) and protein expression (Western blotting) of PPARα, PPARδ, and PPARγ, and on mRNA expression of RXRα, RXRβ, and RXRγ, in bovine epithelial, stromal, and myometrial primary uterine cells and in bovine stromal cells with silenced PPAR genes (N = 10). All PPAR and RXR isoforms were expressed. Prostaglandins affected expression of PPARs only in stromal cells, whereas LTs modulated PPARγ mRNA expression in epithelial and myometrial primary cells. Blockade of signal transduction through PPARs prevented interactions between AA metabolites and PPARs and changed RXR expression comparing with primary stromal cells. In primary stromal uterine cells, mRNA expression of RXRs was higher than that of PPARs. In uterine stromal cells in which intracellular signaling through PPARs was blocked, RXRs seem to take over the role of PPARs and are pivotal for cell functions. This study revealed the reaction of PPARs and RXRs to agonists which naturally occur in the bovine uterus.
Collapse
Affiliation(s)
- A A Szczepańska
- Department of Biodiversity Protection, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Olsztyn, Tuwima St. 10, 10-747 Olsztyn, Poland
| | - M Łupicka
- Department of Biodiversity Protection, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Olsztyn, Tuwima St. 10, 10-747 Olsztyn, Poland
| | - B M Socha
- Department of Biodiversity Protection, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Olsztyn, Tuwima St. 10, 10-747 Olsztyn, Poland
| | - A J Korzekwa
- Department of Biodiversity Protection, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Olsztyn, Tuwima St. 10, 10-747 Olsztyn, Poland. http://tbr.pan.olsztyn.pl/images/KSIAZKA_KONFERENCYJNA_TBR_30_08_17.pdf
| |
Collapse
|
8
|
Bandaletova TY. Morphogenesis of Epithelial Lesions in the Lymphoid Patches of Fischer 344 Rats during Cyclosporine-A-Induced Prolonged Immunosuppression. TUMORI JOURNAL 2018; 78:366-9. [PMID: 1297229 DOI: 10.1177/030089169207800603] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Alterations of intestinal lymphoid patches induced by cyclosporine A (CS-A) were studied in male Fischer 344 rats. Continuous treatment with CS-A (10 mg/kg b.w. by gavage daily) resulted in lymphocyte deficiency of the intestinal lymphoid patches followed by progressive replacement of the lymphoid tissue by cystic and glandular epithelial structures and single cells positive for epithelium-associated immunohistochemical markers. Cessation of CS-A administration led to regression of the alterations and a moderate recovery of the lymphoid patches. Morphologic changes induced at the epithelial-lymphoid border may be a useful parameter to estimate immunotoxicity.
Collapse
|
9
|
Di Domenico M, Giordano A. Signal transduction growth factors: the effective governance of transcription and cellular adhesion in cancer invasion. Oncotarget 2018; 8:36869-36884. [PMID: 28415812 PMCID: PMC5482705 DOI: 10.18632/oncotarget.16300] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 03/01/2017] [Indexed: 12/15/2022] Open
Abstract
Giulio Bizzozero classified the tissues concerning their capacity to self-renew during the adult life in labile, stable and permanent tissues. In 1940 Viktor Hamburger and Rita Levi Montalcini exposed the possibility to induce the growth of permanent cells thanks to a specific ligand Nerve Growth Factor (NGF). Stanley Cohen purified a protein the Epidermal Growth Factor (EGF), able to induce epidermis proliferation and to elicit precocious eye disclosure and teeth eruption, establishing the “inverse” relationships between the proliferation and differentiation. These two biological effects induced by EGF were according to EGFR signaling is involved in a large array of cellular functions such as proliferation, survival, adhesion, migration and differentiation. This review is focused on the key role of growth factors signaling and their downstream effectors in physiological and in pathological phenomena, the authors highlight the governance of Growth factors during the EMT in cancer invasion.
Collapse
Affiliation(s)
- Marina Di Domenico
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Italy.,IRCCS Institute of Women's Health Malzoni Clinic, Avellino, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA, USA
| | - Antonio Giordano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA, USA
| |
Collapse
|
10
|
Kaye PM. Stromal Cell Responses in Infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1060:23-36. [DOI: 10.1007/978-3-319-78127-3_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
11
|
Stromal Clues in Endometrial Carcinoma: Loss of Expression of β-Catenin, Epithelial-Mesenchymal Transition Regulators, and Estrogen-Progesterone Receptor. Int J Gynecol Pathol 2017; 35:238-48. [PMID: 26367784 PMCID: PMC4823869 DOI: 10.1097/pgp.0000000000000233] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Supplemental Digital Content is available in the text. Epithelial-stroma interactions in the endometrium are known to be responsible for physiological functions and emergence of several pathologic lesions. Periglandular stromal cells act on endometrial cells in a paracrine manner through sex hormones. In this study, we immunohistochemically evaluated the expression of epithelial-mesenchymal transition regulators (SNAIL/SLUG, TWIST, ZEB1), adhesion molecules (β-catenin and E-cadhenin), estrogen (ER)-progesterone (PR) receptor and their correlation with each other in 30 benign, 148 hyperplastic (EH), and 101 endometrioid-type endometrial carcinoma (EC) endometria. In the epithelial component, loss of expression in E-cadherin, ER and PR, and overexpression of TWIST and ZEB1 were significantly higher in EC than in EH (P<0.01). In the periglandular stromal component, β-catenin and SNAIL/SLUG expression were significantly higher in normal endometrium and simple without atypical EH compared with complex atypical EH and EC (P<0.01). In addition, periglandular stromal TWIST expression was significantly higher in EH group compared with EC (P<0.05). There was significantly negative correlation between β-catenin and ER, TWIST and ER, and TWIST and PR in hyperplastic and carcinomatous glandular epithelium, whereas there was a significantly positive correlation between β-catenin and SNAIL-SLUG, β-catenin and TWIST, β-catenin and ER, β-catenin and PR, SNAIL-SLUG and ER, SNAIL-SLUG and PR, TWIST and ER, TWIST and PR, in periglandular/cancer-associated stromal cells (P<0.01). In conclusion, the pattern of positive and negative correlations in the expression of epithelial-mesenchymal transition regulators (SNAIL-SLUG and TWIST), sex hormone receptors (ER and PR), and β-catenin between ECs and hyperplasia, as well as between epithelium and stroma herein, is suggestive of a significant role for these proteins and their underlying molecular processes in the development of endometrial carcinomas.
Collapse
|
12
|
Hindman AR, Mo XM, Helber HL, Kovalchin CE, Ravichandran N, Murphy AR, Fagan AM, St. John PM, Burd CJ. Varying Susceptibility of the Female Mammary Gland to In Utero Windows of BPA Exposure. Endocrinology 2017; 158:3435-3447. [PMID: 28938483 PMCID: PMC5659685 DOI: 10.1210/en.2017-00116] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 07/31/2017] [Indexed: 11/19/2022]
Abstract
In utero exposure to the endocrine disrupting compound bisphenol A (BPA) is known to disrupt mammary gland development and increase tumor susceptibility in rodents. It is unclear whether different periods of in utero development might be more susceptible to BPA exposure. We exposed pregnant CD-1 mice to BPA at different times during gestation that correspond to specific milestones of in utero mammary gland development. The mammary glands of early-life and adult female mice, exposed in utero to BPA, were morphologically and molecularly (estrogen receptor-α and Ki67) evaluated for developmental abnormalities. We found that BPA treatment occurring before mammary bud invasion into the mesenchyme [embryonic day (E)12.5] incompletely resulted in the measured phenotypes of mammary gland defects. Exposing mice up to the point at which the epithelium extends into the precursor fat pad (E16.5) resulted in a nearly complete BPA phenotype and exposure during epithelial extension (E15.5 to E18.5) resulted in a partial phenotype. Furthermore, the relative differences in phenotypes between exposure windows highlight the substantial correlations between early-life molecular changes (estrogen receptor-α and Ki67) in the stroma and the epithelial elongation defects in mammary development. These data further implicate BPA action in the stroma as a critical mediator of epithelial phenotypes.
Collapse
Affiliation(s)
- Andrea R. Hindman
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio 43210
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210
| | - Xiaokui Molly Mo
- Center for Biostatistics, The Ohio State University Wexner Medical Center, Columbus, Ohio 43210
| | - Hannah L. Helber
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio 43210
| | - Claire E. Kovalchin
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio 43210
| | | | - Alina R. Murphy
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio 43210
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210
| | - Abigail M. Fagan
- Department of Chemistry, State University of New York New Paltz, New Paltz, New York 12561
| | - Pamela M. St. John
- Department of Chemistry, State University of New York New Paltz, New Paltz, New York 12561
| | - Craig J. Burd
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio 43210
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210
| |
Collapse
|
13
|
Guszczyn T, Surażyński A, Zaręba I, Rysiak E, Popko J, Pałka J. Differential effect of platelet-rich plasma fractions on β1-integrin signaling, collagen biosynthesis, and prolidase activity in human skin fibroblasts. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:1849-1857. [PMID: 28694685 PMCID: PMC5491703 DOI: 10.2147/dddt.s135949] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The study was conducted to evaluate the effects of platelet-rich plasma (PRP), supernatant of PRP (SPRP) obtained by centrifugation, and supernatant of activated PRP (SActi-PRP) obtained by Ca2+ solution-treated PRP on collagen biosynthesis, prolidase activity, and β1-integrin signaling in cultured human skin fibroblasts. Incubation of fibroblasts with 5% PRP for 24 h contributed to ~5-fold increase in collagen biosynthesis compared to the control. In the cells treated with 5% of SPRP or SActi-PRP, collagen biosynthesis showed a 3-fold increase of the control. PRP, SPRP, and SActi-PRP stimulated prolidase activity similar to collagen biosynthesis. Collagen biosynthesis and prolidase activity are regulated by β1-integrin receptor signaling. Incubation of fibroblasts with PRP for 24 h contributed to a dose-dependent increase in the expression of β1-integrin receptor, while SActi-PRP increased the process to a much lower extent. SPRP had no effect on the β1-integrin receptor expression. All the studied fractions of blood increased the expression of FAK as well as the expression of phosphorylated MAP-kinases. However, PRP was found to be the most effective stimulator of expression of these particular kinases. These studies suggest that a complex of factors, including growth factors, adhesion molecules, and prolidase contained in PRP, all evoke growth and collagen-promoting activities in human dermal fibroblasts.
Collapse
Affiliation(s)
| | - Arkadiusz Surażyński
- Department of Medicinal Chemistry, Medical University of Białystok, Białystok, Poland
| | - Ilona Zaręba
- Department of Medicinal Chemistry, Medical University of Białystok, Białystok, Poland
| | - Edyta Rysiak
- Department of Medicinal Chemistry, Medical University of Białystok, Białystok, Poland
| | - Janusz Popko
- Department of Pediatric Orthopedics and Traumatology
| | - Jerzy Pałka
- Department of Medicinal Chemistry, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
14
|
RhoA knockout fibroblasts lose tumor-inhibitory capacity in vitro and promote tumor growth in vivo. Proc Natl Acad Sci U S A 2017; 114:E1413-E1421. [PMID: 28174275 DOI: 10.1073/pnas.1621161114] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Fibroblasts are a main player in the tumor-inhibitory microenvironment. Upon tumor initiation and progression, fibroblasts can lose their tumor-inhibitory capacity and promote tumor growth. The molecular mechanisms that underlie this switch have not been defined completely. Previously, we identified four proteins overexpressed in cancer-associated fibroblasts and linked to Rho GTPase signaling. Here, we show that knocking out the Ras homolog family member A (RhoA) gene in normal fibroblasts decreased their tumor-inhibitory capacity, as judged by neighbor suppression in vitro and accompanied by promotion of tumor growth in vivo. This also induced PC3 cancer cell motility and increased colony size in 2D cultures. RhoA knockout in fibroblasts induced vimentin intermediate filament reorganization, accompanied by reduced contractile force and increased stiffness of cells. There was also loss of wide F-actin stress fibers and large focal adhesions. In addition, we observed a significant loss of α-smooth muscle actin, which indicates a difference between RhoA knockout fibroblasts and classic cancer-associated fibroblasts. In 3D collagen matrix, RhoA knockout reduced fibroblast branching and meshwork formation and resulted in more compactly clustered tumor-cell colonies in coculture with PC3 cells, which might boost tumor stem-like properties. Coculturing RhoA knockout fibroblasts and PC3 cells induced expression of proinflammatory genes in both. Inflammatory mediators may induce tumor cell stemness. Network enrichment analysis of transcriptomic changes, however, revealed that the Rho signaling pathway per se was significantly triggered only after coculturing with tumor cells. Taken together, our findings in vivo and in vitro indicate that Rho signaling governs the inhibitory effects by fibroblasts on tumor-cell growth.
Collapse
|
15
|
Zhang H, Li H, Qi S, Liu Z, Fu Y, Li M, Zhao X. Normal endometrial stromal cells regulate 17β-estradiol-induced epithelial-mesenchymal transition via slug and E-cadherin in endometrial adenocarcinoma cells in vitro. Gynecol Endocrinol 2017; 33:82-86. [PMID: 27449470 DOI: 10.1080/09513590.2016.1203896] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Stroma-tumor communication participates in the pathogenesis of endometrial carcinomas. In previous studies, we found that normal stromal cells inhibited the growth of endometrial carcinoma cells. Here, we investigated the role of normal stromal cells in the epithelial-mesenchymal transition (EMT) of endometrial carcinoma cells and explored the possible mechanism implied. We found that conditioned medium (CM) by normal endometrial stromal cells (NSC) reduced cell growth and induced cell apoptosis in Ishikawa cells. CM by NSC inhibited 17β-estradiol-induced cell growth and apoptosis decrease in Ishikawa cells. Moreover, CM by NSC inhibited the migration and invasion, and 17β-estradiol-induced migration and invasion in Ishikawa cells. Meanwhile, CM by NSC decreased Slug expression and 17β-estradiol-induced Slug expression, increased E-cadherin expression and abolished 17β-estradiol-induced E-cadherin reduction in Ishikawa cells. In conclusion, normal stromal factors can inhibit 17β-estradiol-induced cell proliferation and apoptosis inhibition, and abolished 17β-estradiol-induced EMT in endometrial cancer cell via regulating E-cadherin and Slug expression.
Collapse
Affiliation(s)
- Hui Zhang
- a Department of Obstetrics and Gynecology and
| | - Hongyan Li
- a Department of Obstetrics and Gynecology and
| | - Shasha Qi
- a Department of Obstetrics and Gynecology and
| | - Zhao Liu
- b Department of Urology , Shandong Provincial Hospital Affiliated to Shandong University , Jinan , Shandong , China
| | - Yibing Fu
- a Department of Obstetrics and Gynecology and
| | | | - Xingbo Zhao
- a Department of Obstetrics and Gynecology and
| |
Collapse
|
16
|
Spatiotemporal dynamics of androgen signaling underlie sexual differentiation and congenital malformations of the urethra and vagina. Proc Natl Acad Sci U S A 2016; 113:E7510-E7517. [PMID: 27821748 DOI: 10.1073/pnas.1610471113] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Disorders of sex development (DSDs) are congenital anomalies that affect sexual differentiation of genitourinary organs and secondary sex characters. A common cause of female genital virilization is congenital adrenal hyperplasia (CAH), in which excess androgen production during development of 46XX females can result in vaginal atresia, masculinization of the urethra, a single urogenital sinus, and clitoral hypertrophy or ambiguous external genitalia. Development of the vagina depends on sexual differentiation of the urogenital sinus ridge, an epithelial thickening that forms where the sex ducts attach to the anterior urethra. In females, the sinus ridge descends posteriorly to allow the vaginal opening to form in the vulva, whereas in males and in females with CAH, androgens inhibit descent of the sinus ridge. The mechanisms that regulate development of the female urethra and vagina are largely unknown. Here we show that the timing and duration of, and the cell population targeted by, androgen signaling determine the position of vaginal attachment to the urethra. Manipulations of androgen signaling in utero reveal a temporal window of development when sinus ridge fate is determined. Cell type-specific genetic deletions of androgen receptor (Ar) identify a subpopulation of mesenchymal cells that regulate sinus ridge morphogenesis. These results reveal a common mechanism that coordinates development of the vagina and feminization of the urethra, which may account for development of a single urogenital sinus in females exposed to excessive androgen during a critical period of prenatal development.
Collapse
|
17
|
Zhang H, Xue J, Li M, Zhao X, Wei D, Li C. Metformin regulates stromal-epithelial cells communication via Wnt2/β-catenin signaling in endometriosis. Mol Cell Endocrinol 2015; 413:61-5. [PMID: 26116230 DOI: 10.1016/j.mce.2015.06.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Revised: 05/30/2015] [Accepted: 06/10/2015] [Indexed: 11/25/2022]
Abstract
In previous studies, we found that endometriotic stromal cells lose the ability to regulate cell survival signaling in endometriotic epithelial cells. Here, we invested the effect of Metformin on the stromal-epithelial cells crosstalk in endometriosis and explored the pathway that might be involved. We found that ectopic endometriotic stromal cells (ESC) expressed and secreted higher Wnt2 protein compared with normal endometrial stromal cells (NSC). Conditioned medium (CM) from ESC supplemented with Wnt2 antibody significantly inhibited the growth of normal endometrial epithelial cells (NEC), while CM from ESC per se showed no significant effect on the growth of NEC. Metformin decreased the expression and secretion of Wnt2 in ESC. CM from Metformin-pretreated ESC significantly inhibited the growth of NEC. In conclusion, Wnt2/β-catenin signaling was involved in stromal-epithelial cells interaction in endometriosis. Metformin might regulate the stroma-epithelium communication via Wnt2-mediated signaling in endometriosis.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, Shandong 250021, People's Republic of China
| | - Jing Xue
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, Shandong 250021, People's Republic of China
| | - Mingjiang Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, Shandong 250021, People's Republic of China
| | - Xingbo Zhao
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, Shandong 250021, People's Republic of China
| | - Deying Wei
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, Shandong 250021, People's Republic of China
| | - Changzhong Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, Shandong 250021, People's Republic of China.
| |
Collapse
|
18
|
Dermal fibroblast expression of stromal cell-derived factor-1 (SDF-1) promotes epidermal keratinocyte proliferation in normal and diseased skin. Protein Cell 2015; 6:890-903. [PMID: 26296527 PMCID: PMC4656211 DOI: 10.1007/s13238-015-0198-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 07/16/2015] [Indexed: 01/24/2023] Open
Abstract
Stromal cells provide a crucial microenvironment for overlying epithelium. Here we investigated the expression and function of a stromal cell-specific protein, stromal cell-derived factor-1 (SDF-1), in normal human skin and in the tissues of diseased skin. Immunohistology and laser capture microdissection (LCM)-coupled quantitative real-time RT-PCR revealed that SDF-1 is constitutively and predominantly expressed in dermal stromal cells in normal human skin in vivo. To our surprise, an extremely high level of SDF-1 transcription was observed in the dermis of normal human skin in vivo, evidenced by much higher mRNA expression level than type I collagen, the most abundant and highly expressed protein in human skin. SDF-1 was also upregulated in the tissues of many human skin disorders including psoriasis, basal cell carcinoma (BCC), and squamous cell carcinoma (SCC). Double immunostaining for SDF-1 and HSP47 (heat shock protein 47), a marker of fibroblasts, revealed that fibroblasts were the major source of stroma-cell-derived SDF-1 in both normal and diseased skin. Functionally, SDF-1 activates the ERK (extracellular-signal-regulated kinases) pathway and functions as a mitogen to stimulate epidermal keratinocyte proliferation. Both overexpression of SDF-1 in dermal fibroblasts and treatment with rhSDF-1 to the skin equivalent cultures significantly increased the number of keratinocyte layers and epidermal thickness. Conversely, the stimulative function of SDF-1 on keratinocyte proliferation was nearly completely eliminated by interfering with CXCR4, a specific receptor of SDF-1, or by knock-down of SDF-1 in fibroblasts. Our data reveal that extremely high levels of SDF-1 provide a crucial microenvironment for epidermal keratinocyte proliferation in both physiologic and pathologic skin conditions.
Collapse
|
19
|
Jerman UD, Kreft ME, Veranič P. Epithelial-Mesenchymal Interactions in Urinary Bladder and Small Intestine and How to Apply Them in Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:521-30. [PMID: 26066408 DOI: 10.1089/ten.teb.2014.0678] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Reciprocal interactions between the epithelium and mesenchyme are essential for the establishment of proper tissue morphology during organogenesis and tissue regeneration as well as for the maintenance of cell differentiation. With this review, we highlight the importance of epithelial-mesenchymal cross talk in healthy tissue and further discuss its significance in engineering functional tissues in vitro. We focus on the urinary bladder and small intestine, organs that are often compromised by disease and are as such in need of research that would advance effective treatment or tissue replacement. To date, the understanding of epithelial-mesenchymal reciprocal interactions has enabled the development of in vitro biomimetic tissue equivalents that have provided many possibilities in treating defective, damaged, or even cancerous tissues. Although research of the past several years has advanced the field of bladder and small intestine tissue engineering, one must be aware of its current limitations in successfully and above all safely introducing tissue-engineered constructs into clinical practice. Special attention is in particular needed when treating cancerous tissues, as initially successful tumor excision and tissue reconstruction may later on result in cancer recurrence due to oncogenic signals originating from an altered stroma. Recent rather poor outcomes in pioneering clinical trials of bladder reconstructions should serve as a reminder that recreating a functional organ to replace a dysfunctional one is an objective far more difficult to reach than initially foreseen. When considering effective tissue engineering approaches for diseased tissues in humans, it is imperative to introduce animal models with dysfunctional or, even more importantly, cancerous organs, which would greatly contribute to predicting possible complications and, hence, reducing risks when translating to the clinic.
Collapse
Affiliation(s)
- Urška Dragin Jerman
- Faculty of Medicine, Institute of Cell Biology, University of Ljubljana , Ljubljana, Slovenia
| | - Mateja Erdani Kreft
- Faculty of Medicine, Institute of Cell Biology, University of Ljubljana , Ljubljana, Slovenia
| | - Peter Veranič
- Faculty of Medicine, Institute of Cell Biology, University of Ljubljana , Ljubljana, Slovenia
| |
Collapse
|
20
|
Abstract
Cancer metastasis is highly inefficient and complex. Common features of metastatic cancer cells have been observed using cancer cell lines and genetically reconstituted mouse and human tumor xenograft models. These include cancer cell interaction with the tumor microenvironment and the ability of cancer cells to sense extracellular stimuli and adapt to adverse growth conditions. This review summarizes the coordinated response of cancer cells to soluble growth factors, such as RANKL, by a unique feed forward mechanism employing coordinated upregulation of RANKL and c-Met with downregulation of androgen receptor. The RANK-mediated signal network was found to drive epithelial to mesenchymal transition in prostate cancer cells, promote osteomimicry and the ability of prostate cancer cells to assume stem cell and neuroendocrine phenotypes, and confer the ability of prostate cancer cells to home to bone. Prostate cancer cells with activated RANK-mediated signal network were observed to recruit and even transform the non-tumorigenic prostate cancer cells to participate in bone and soft tissue colonization. The coordinated regulation of cancer cell invasion and metastasis by the feed forward mechanism involving RANKL, c-Met, transcription factors, and VEGF-neuropilin could offer new therapeutic opportunities to target prostate cancer bone and soft tissue metastases.
Collapse
Affiliation(s)
- Gina Chia-Yi Chu
- Departments of Medicine and Surgery, Samuel Orchin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA,
| | | |
Collapse
|
21
|
Zhang P, Lo A, Huang Y, Huang G, Liang G, Mott J, Karpen GH, Blakely EA, Bissell MJ, Barcellos-Hoff MH, Snijders AM, Mao JH. Identification of genetic loci that control mammary tumor susceptibility through the host microenvironment. Sci Rep 2015; 5:8919. [PMID: 25747469 PMCID: PMC4352890 DOI: 10.1038/srep08919] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 01/15/2015] [Indexed: 11/25/2022] Open
Abstract
The interplay between host genetics, tumor microenvironment and environmental exposure in cancer susceptibility remains poorly understood. Here we assessed the genetic control of stromal mediation of mammary tumor susceptibility to low dose ionizing radiation (LDIR) using backcrossed F1 into BALB/c (F1Bx) between cancer susceptible (BALB/c) and resistant (SPRET/EiJ) mouse strains. Tumor formation was evaluated after transplantation of non-irradiated Trp53-/- BALB/c mammary gland fragments into cleared fat pads of F1Bx hosts. Genome-wide linkage analysis revealed 2 genetic loci that constitute the baseline susceptibility via host microenvironment. However, once challenged with LDIR, we discovered 13 additional loci that were enriched for genes involved in cytokines, including TGFβ1 signaling. Surprisingly, LDIR-treated F1Bx cohort significantly reduced incidence of mammary tumors from Trp53-/- fragments as well as prolonged tumor latency, compared to sham-treated controls. We demonstrated further that plasma levels of specific cytokines were significantly correlated with tumor latency. Using an ex vivo 3-D assay, we confirmed TGFβ1 as a strong candidate for reduced mammary invasion in SPRET/EiJ, which could explain resistance of this strain to mammary cancer risk following LDIR. Our results open possible new avenues to understand mechanisms of genes operating via the stroma that affect cancer risk from external environmental exposures.
Collapse
Affiliation(s)
- Pengju Zhang
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Alvin Lo
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Yurong Huang
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Ge Huang
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Guozhou Liang
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Joni Mott
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Gary H. Karpen
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Eleanor A. Blakely
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Mina J. Bissell
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | | | - Antoine M. Snijders
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Jian-Hua Mao
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| |
Collapse
|
22
|
An analytical method for the quantification of hERG1 channel gene expression in human colorectal cancer. ACTA ACUST UNITED AC 2014; 22:215-21. [PMID: 24193004 DOI: 10.1097/pdm.0b013e31828e55c7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cancer molecular investigation revealed a huge molecular heterogeneity between different types of cancers as well as among cancer patients affected by the same cancer type. This implies the necessity of a personalized approach for cancer diagnosis and therapy, on the basis of the development of standardized protocols to facilitate the application of molecular techniques in the clinical decision-making process. Ion channels encoding genes are acquiring increasing relevance in oncological translational studies, representing new candidates for molecular diagnostic and therapeutic purposes. Hence, the development of molecular protocols for the quantification of ion channels encoding genes in tumor specimens may have relevance for diagnostic and prognostic investigation. Two main hindrances must be overcome for these purposes: the use of formalin-fixed and paraffin-embedded samples for gene expression analysis and the physiological expression of ion channels in excitable cells, potentially present in the tumor sample. We here propose a method for hERG1 gene quantification in colorectal cancer samples in both cryopreserved and formalin-fixed and paraffin-embedded samples. An analytical method was developed to estimate hERG1 gene expression exclusively in epithelial cancer cells. Indeed, we found that the hERG1 gene was expressed at significant levels by myofibroblasts present in the tumor stroma. This method was based on the normalization on a smooth muscle-myofibroblast-specific gene, MYH11, with no need of microdissection. By applying this method, hERG1 expression turned out to correlate with VEGF-A expression, confirming previous immunohistochemical data.
Collapse
|
23
|
Yu M, Finner A, Shapiro J, Lo B, Barekatain A, McElwee KJ. Hair follicles and their role in skin health. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/17469872.1.6.855] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
24
|
Heterogeneity of functional properties of Clone 66 murine breast cancer cells expressing various stem cell phenotypes. PLoS One 2013; 8:e78725. [PMID: 24265713 PMCID: PMC3827106 DOI: 10.1371/journal.pone.0078725] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 09/19/2013] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION Breast cancer grows, metastasizes and relapses from rare, therapy resistant cells with a stem cell phenotype (cancer stem cells/CSCs). However, there is a lack of studies comparing the functions of CSCs isolated using different phenotypes in order to determine if CSCs are homogeneous or heterogeneous. METHODS Cells with various stem cell phenotypes were isolated by sorting from Clone 66 murine breast cancer cells that grow orthotopically in immune intact syngeneic mice. These populations were compared by in vitro functional assays for proliferation, growth, sphere and colony formation; and in vivo limiting dilution analysis of tumorigenesis. RESULTS The proportion of cells expressing CD44(high)CD24(low/neg), side population (SP) cells, ALDH1(+), CD49f(high), CD133(high), and CD34(high) differed, suggesting heterogeneity. Differences in frequency and size of tumor spheres from these populations were observed. Higher rates of proliferation of non-SP, ALDH1(+), CD34(low), and CD49f(high) suggested properties of transit amplifying cells. Colony formation was higher from ALDH1(-) and non-SP cells than ALDH1(+) and SP cells suggesting a progenitor phenotype. The frequency of clonal colonies that grew in agar varied and was differentially altered by the presence of Matrigel™. In vivo, fewer cells with a stem cell phenotype were needed for tumor formation than "non-stem" cells. Fewer SP cells were needed to form tumors than ALDH1(+) cells suggesting further heterogeneities of cells with stem phenotypes. Different levels of cytokines/chemokines were produced by Clone 66 with RANTES being the highest. Whether the heterogeneity reflects soluble factor production remains to be determined. CONCLUSIONS These data demonstrate that Clone 66 murine breast cancer cells that express stem cell phenotypes are heterogeneous and exhibit different functional properties, and this may also be the case for human breast cancer stem cells.
Collapse
|
25
|
Subramaniam KS, Tham ST, Mohamed Z, Woo YL, Mat Adenan NA, Chung I. Cancer-associated fibroblasts promote proliferation of endometrial cancer cells. PLoS One 2013; 8:e68923. [PMID: 23922669 PMCID: PMC3724864 DOI: 10.1371/journal.pone.0068923] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 06/03/2013] [Indexed: 01/25/2023] Open
Abstract
Endometrial cancer is the most commonly diagnosed gynecologic malignancy worldwide; yet the tumor microenvironment, especially the fibroblast cells surrounding the cancer cells, is poorly understood. We established four primary cultures of fibroblasts from human endometrial cancer tissues (cancer-associated fibroblasts, CAFs) using antibody-conjugated magnetic bead isolation. These relatively homogenous fibroblast cultures expressed fibroblast markers (CD90, vimentin and alpha-smooth muscle actin) and hormonal (estrogen and progesterone) receptors. Conditioned media collected from CAFs induced a dose-dependent proliferation of both primary cultures and cell lines of endometrial cancer in vitro (175%) when compared to non-treated cells, in contrast to those from normal endometrial fibroblast cell line (51%) (P<0.0001). These effects were not observed in fibroblast culture derived from benign endometrial hyperplasia tissues, indicating the specificity of CAFs in affecting endometrial cancer cell proliferation. To determine the mechanism underlying the differential fibroblast effects, we compared the activation of PI3K/Akt and MAPK/Erk pathways in endometrial cancer cells following treatment with normal fibroblasts- and CAFs-conditioned media. Western blot analysis showed that the expression of both phosphorylated forms of Akt and Erk were significantly down-regulated in normal fibroblasts-treated cells, but were up-regulated/maintained in CAFs-treated cells. Treatment with specific inhibitors LY294002 and U0126 reversed the CAFs-mediated cell proliferation (P<0.0001), suggesting for a role of these pathways in modulating endometrial cancer cell proliferation. Rapamycin, which targets a downstream molecule in PI3K pathway (mTOR), also suppressed CAFs-induced cell proliferation by inducing apoptosis. Cytokine profiling analysis revealed that CAFs secrete higher levels of macrophage chemoattractant protein (MCP)-1, interleukin (IL)-6, IL-8, RANTES and vascular endothelial growth factor (VEGF) than normal fibroblasts. Our data suggests that in contrast to normal fibroblasts, CAFs may exhibit a pro-tumorigenic effect in the progression of endometrial cancer, and PI3K/Akt and MAPK/Erk signaling may represent critical regulators in how endometrial cancer cells respond to their microenvironment.
Collapse
Affiliation(s)
- Kavita S. Subramaniam
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- University of Malaya Cancer Research Institute, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Seng Tian Tham
- Department of Obstetrics & Gynecology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- University of Malaya Cancer Research Institute, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Zahurin Mohamed
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Pharmacogenomics Laboratory, Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yin Ling Woo
- Department of Obstetrics & Gynecology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- University of Malaya Cancer Research Institute, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Noor Azmi Mat Adenan
- Department of Obstetrics & Gynecology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Ivy Chung
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- University of Malaya Cancer Research Institute, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- * E-mail:
| |
Collapse
|
26
|
Jacca S, Franceschi V, Colagiorgi A, Sheldon M, Donofrio G. Bovine Endometrial Stromal Cells Support Tumor Necrosis Factor Alpha-Induced Bovine Herpesvirus Type 4 Enhanced Replication1. Biol Reprod 2013; 88:135. [DOI: 10.1095/biolreprod.112.106740] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
27
|
D’Amici S, Ceccarelli S, Vescarelli E, Romano F, Frati L, Marchese C, Angeloni A. TNFα modulates Fibroblast Growth Factor Receptor 2 gene expression through the pRB/E2F1 pathway: identification of a non-canonical E2F binding motif. PLoS One 2013; 8:e61491. [PMID: 23613863 PMCID: PMC3629046 DOI: 10.1371/journal.pone.0061491] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 03/08/2013] [Indexed: 11/18/2022] Open
Abstract
Interactions between epithelium and mesenchyme during wound healing are not fully understood, but Fibroblast Growth Factors (FGFs) and their receptors FGFRs are recognized as key elements. FGFR2 gene encodes for two splicing transcript variants, FGFR2-IIIb or Keratinocyte Growth Factor Receptor (KGFR) and FGFR2-IIIc, which differ for tissue localization and ligand specificity. Proinflammatory cytokines play an essential role in the regulation of epithelial-mesenchymal interactions, and have been indicated to stimulate FGFs production. Here we demonstrated that upregulation of FGFR2 mRNA and protein expression is induced by the proinflammatory cytokines Tumor Necrosis Factor-α, Interleukin-1β and Interleukin 2. Furthermore, we found that TNFα determines FGFR2 transcriptional induction through activation of pRb, mediated by Raf and/or p38 pathways, and subsequent release of the transcription factor E2F1. Experiments based on FGFR2 promoter serial deletions and site-directed mutagenesis allowed us to identify a minimal responsive element that retains the capacity to be activated by E2F1. Computational analysis indicated that this element is a non-canonical E2F responsive motif. Thus far, the molecular mechanisms of FGFR2 upregulation during wound healing or in pathological events are not known. Our data suggest that FGFR2 expression can be modulated by local recruitment of inflammatory cytokines. Furthermore, since alterations in FGFR2 expression have been linked to the pathogenesis of certain human cancers, these findings could also provide elements for diagnosis and potential targets for novel therapeutic approaches.
Collapse
Affiliation(s)
- Sirio D’Amici
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Simona Ceccarelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Enrica Vescarelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Ferdinando Romano
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Luigi Frati
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Cinzia Marchese
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Antonio Angeloni
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- * E-mail:
| |
Collapse
|
28
|
MacKintosh SB, Schuberth HJ, Healy LL, Sheldon IM. Polarised bovine endometrial epithelial cells vectorially secrete prostaglandins and chemotactic factors under physiological and pathological conditions. Reproduction 2013; 145:57-72. [PMID: 23115348 DOI: 10.1530/rep-12-0253] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Epithelial cells of the endometrium secrete prostaglandins to regulate the bovine oestrous cycle and form a functional barrier to microbes. However, bacterial infection of the endometrium commonly causes infertility in dairy cattle by disrupting endometrial physiology. Epithelial cell cultures are used to study the mechanisms of physiology and pathology, but 2D cultures may not reflect the 3D complexity of the epithelium. In this study, a polarised epithelial cell transwell culture was developed, using transepithelial resistance (TER), to monitor epithelial integrity. Polarised epithelial cells were treated with oxytocin and arachidonic acid to test physiological function and with lipopolysaccharide (LPS) to mimic bacterial infection. Supernatants were analysed for prostaglandin E(2) (PGE), prostaglandin F(2)(α), the chemokine interleukin-8 (IL8) and the ability of supernatants to induce neutrophil migration. Confluent epithelial cells established polarity when TER was >1800 Ω cm(2) and predominantly released prostaglandins basolaterally. In contrast, IL8 from epithelial cells accumulated apically and the supernatants were highly chemotactic for neutrophils. The striking exception was when the epithelial cells were treated with LPS in the apical or basolateral compartment independently, which led to the release of IL8 towards the treated compartment. Although stromal cells also accumulated PGE and IL8 in response to treatment, co-culture of stromal cells in the well below polarised epithelial cells did not influence cellular responses. In conclusion, polarised endometrial epithelial cells vectorially released prostaglandins and chemokines to reflect their respective mechanistic roles in physiology and pathology.
Collapse
Affiliation(s)
- Siân B MacKintosh
- School of Medicine, College of Medicine, Institute of Life Science, Swansea University, Singleton Park, Swansea SA2 8PP, UK.
| | | | | | | |
Collapse
|
29
|
Watnick RS. The role of the tumor microenvironment in regulating angiogenesis. Cold Spring Harb Perspect Med 2012; 2:a006676. [PMID: 23209177 DOI: 10.1101/cshperspect.a006676] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The tumor-associated stroma has been shown to play a significant role in cancer formation. Paracrine signaling interactions between epithelial tumor cells and stromal cells are a key component in the transformation and proliferation of tumors in several organs. Whereas the intracellular signaling pathways regulating the expression of several pro- and antiangiogenic proteins have been well characterized in human cancer cells, the intercellular signaling that takes place between tumor cells and the surrounding tumor-associated stroma has not been as extensively studied with regard to the regulation of angiogenesis. In this chapter we define the key players in the regulation of angiogenesis and examine how their expression is regulated in the tumor-associated stroma. The resulting analysis is often seemingly paradoxical, underscoring the complexity of intercellular signaling within tumors and the need to better understand the environmental context underlying these signaling mechanisms.
Collapse
Affiliation(s)
- Randolph S Watnick
- Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
30
|
Survivin, MMP-2, MT1-MMP, and TIMP-2: their impact on survival, implantation, and proliferation of endometriotic tissues. Virchows Arch 2012; 461:589-99. [DOI: 10.1007/s00428-012-1301-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 08/09/2012] [Accepted: 08/09/2012] [Indexed: 02/03/2023]
|
31
|
Silveira CGT, Abrão MS, Dias JA, Coudry RA, Soares FA, Drigo SA, Domingues MAC, Rogatto SR. Common chromosomal imbalances and stemness-related protein expression markers in endometriotic lesions from different anatomical sites: the potential role of stem cells. Hum Reprod 2012; 27:3187-97. [PMID: 22940770 DOI: 10.1093/humrep/des282] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Endometriosis is a multifactorial gynecological disease characterized by the presence of functional endometrium-like tissue in ectopic sites. Several studies have focused on elucidating the immunological, endocrine, environmental and genetic factors involved in endometriosis. However, its pathogenesis is still unclear. METHODS High-resolution comparative genomic hybridization was applied to screen for genomic imbalances in laser microdissected stromal and epithelial cells from 20 endometriotic lesions and three samples of eutopic endometrium derived from eight patients. The expression of seven stemness-related markers (CD9, CD13, CD24, CD34, CD133, CD117/c-Kit and Oct-4) in endometrial tissue samples was evaluated by immunohistochemistry. RESULTS Samples of eutopic endometrium showed normal genomic profiles. In ectopic tissues, an average of 68 genomic imbalances was detected per sample. DNA losses were more frequently detected and involved mainly 3p, 5q, 7p, 9p, 11q, 16q, 18q and 19q. Many of the genomic imbalances detected were common to endometriotic stroma and epithelia and also among different endometriotic sites from the same patient. These findings suggested a clonal origin of the endometriotic cells and the putative involvement of stem cells. Positive immunostaining for CD9, CD34, c-Kit and Oct-4 markers was detected in isolated epithelial and/or stromal cells in eutopic and ectopic endometrium in the majority of cases. CONCLUSIONS The presence of shared genomic alterations in stromal and epithelial cells from different anatomical sites of the same patient and the expression of stemness-related markers suggested that endometriosis arises as a clonal proliferation with the putative involvement of stem cells.
Collapse
Affiliation(s)
- Cássia G T Silveira
- Department of Genetics, Institute of Biosciences, UNESP, São Paulo State University, Botucatu, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Joshi A, Wang H, Jiang G, Douglas W, Chan JSY, Korach KS, Ellenson LH. Endometrial tumorigenesis in Pten(+/-) mice is independent of coexistence of estrogen and estrogen receptor α. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:2536-47. [PMID: 22503752 DOI: 10.1016/j.ajpath.2012.03.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Revised: 01/23/2012] [Accepted: 03/01/2012] [Indexed: 10/28/2022]
Abstract
Numerous studies support the role for mutations in the phosphatase and tensin homologue (PTEN) tumor suppressor gene and unopposed estrogen stimulation in the pathogenesis of uterine endometrioid carcinoma. However, the relation between PTEN signaling and estrogen/estrogen receptor in endometrial tumorigenesis remains unresolved. We used genetically engineered mice as a model to address this relation. Mice with a single deleted Pten allele (Pten(+/-)) spontaneously develop complex atypical hyperplasia and ~20% develop endometrial cancer. To determine the effect of removing endogenous estrogen, we performed oophorectomies on Pten(+/-) mice. Although there was a reduction in the number and severity of hyperplastic lesions, the endometrial phenotype persisted, suggesting that Pten mutation, independent of estrogen, can initiate the development of complex atypical hyperplasia. To recapitulate the situation in women with unopposed estrogen, we implanted 17β-estradiol pellets in adult female Pten heterozygous mice, resulting in increased carcinoma incidence. Because studies have shown that estrogen largely acts on the endometrium via estrogen receptor ERα, we generated Pten(+/-)ERα(-/-) mice. Strikingly, 88.9% of Pten(+/-)ERα(-/-) mice developed endometrial hyperplasia/carcinoma. Furthermore, Pten(+/-)ERα(-/-) mice showed a higher incidence of in situ and invasive carcinoma, suggesting that endometrial tumorigenesis can progress in the absence of ERα. Thus, the relation between Pten alterations and estrogen signaling in the development of endometrial carcinoma is complex; the results presented herein have important implications for the treatment of endometrial hyperplasia and carcinoma in women.
Collapse
Affiliation(s)
- Ayesha Joshi
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Martin PM, Ouafik L. Angiogenèse: retour au fondamental. ONCOLOGIE 2012. [DOI: 10.1007/s10269-012-2152-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
34
|
Shi M, Zhang H, Li M, Xue J, Fu Y, Yan L, Zhao X. Normal endometrial stromal cells regulate survival and apoptosis signaling through PI3K/AKt/Survivin pathway in endometrial adenocarcinoma cells in vitro. Gynecol Oncol 2011; 123:387-92. [PMID: 21794903 DOI: 10.1016/j.ygyno.2011.07.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 06/30/2011] [Accepted: 07/05/2011] [Indexed: 10/17/2022]
Abstract
OBJECTIVE Stroma-tumor communication plays an important role in the genesis of neoplasia. In the current study, we investigated the effect of normal stromal cells on the survival and apoptosis signaling of endometrial cancer cells and further explored the possible mechanism implied in this communication. METHODS Using primarily cultured normal endometrial stromal cells and an endometrial adenocarcinoma cell line, Ishikawa cells, we established a 2D-coculture system to observe the stromal cell-tumor cell crosstalk in endometrial carcinomas. Using methyl thiazolyl tetrazolium (MTT) assays, cell counting and colony formation assays, we analyzed the effect of stomal cells on the growth and proliferation of Ishikawa cells under different conditions. Using western blot analysis, we determined the effect of stromal cells on the activity of PI3K/AKt/Survivin signaling in Ishikawa cells under different conditions. Using immunohistochemistry analysis, we determined the expression of Survivin in normal endometria and endometrial adenocarcinomas. RESULTS We found that the paracrine factors from normal endometrial stromal cells grown on Matrigel repeatedly and significantly decreased hormone-stimulated activity of PI3K/AKt/Survivin signaling in Ishikawa cells, which were proved to be increased in endometrial adenocarcinoma and essential in hormone-induced cell growth in Ishikawa cells. CONCLUSION Paracrine factors from normal endometrial stromal cells can inhibit hormone-stimulated cell proliferation in Ishikawa cells by regulating cell survival and apoptosis through PI3K/AKt/Survivin signaling.
Collapse
Affiliation(s)
- Min Shi
- Department of Obstetrics & Gynecology, Shandong University, Shandong, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
35
|
Haddad SN, Wira CR. Keratinocyte Growth Factor Stimulates Macrophage Inflammatory Protein 3α and Keratinocyte-derived Chemokine Secretion by Mouse Uterine Epithelial Cells. Am J Reprod Immunol 2011; 64:197-211. [PMID: 20455876 DOI: 10.1111/j.1600-0897.2010.00850.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PROBLEM communication between uterine epithelial cells and the underlying stromal fibroblasts is critical for proper endometrial function. Stromal fibroblast-derived growth factors have been shown to regulate epithelial immune functions. The purpose of this study was to determine whether keratinocyte growth factor (KGF) regulates uterine epithelial cell chemokine and antimicrobial secretion. METHOD OF STUDY uterine epithelial cells were isolated from Balb/c mice and cultured in either 96-well plates or transwell inserts. Epithelial cells were treated with KGF, epidermal growth factor (EGF), or hepatocyte growth factor (HGF). Macrophage inflammatory protein 3α (MIP3α) and keratinocyte-derived chemokine (KC) levels were measured by ELISA. RESULTS keratinocyte growth factor stimulated the secretion of MIP3α and KC. The effects on MIP3α by KGF were specific because EGF and HGF had no effect. In contrast, KGF, EGF, and HGF had similar effects on KC. Furthermore, KGF administered to the apical side of epithelial cells had no effect on MIP3α or KC secretion, indicating that the KGF receptor is located on the basolateral surface of uterine epithelial cells. CONCLUSION we demonstrate that KGF plays a role in uterine epithelial cell secretion of MIP3α and KC, key immune mediators involved in the protection of mucosal surfaces in the female reproductive tract.
Collapse
Affiliation(s)
- Severina N Haddad
- Department of Physiology, Dartmouth Medical School, One Medical Center Drive, Lebanon, NH 03756, USA
| | | |
Collapse
|
36
|
Liu M, Xu J, Deng H. Tangled fibroblasts in tumor-stroma interactions. Int J Cancer 2011; 129:1795-805. [DOI: 10.1002/ijc.26116] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Accepted: 03/21/2011] [Indexed: 12/13/2022]
|
37
|
Pérez-Losada J, Castellanos-Martín A, Mao JH. Cancer evolution and individual susceptibility. Integr Biol (Camb) 2011; 3:316-328. [PMID: 21264404 PMCID: PMC3125395 DOI: 10.1039/c0ib00094a] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer susceptibility is due to interactions between inherited genetic factors and exposure to environmental carcinogens. The genetic component is constituted mainly by weakly acting low-penetrance genetic variants that interact among themselves, as well as with the environment. These low susceptibility genes can be categorized into two main groups: one includes those that control intrinsic tumor cell activities (i.e. apoptosis, proliferation or DNA repair), and the other contains those that modulate the function of extrinsic tumor cell compartments (i.e. stroma, angiogenesis, or endocrine and immune systems). Genome-Wide Association Studies (GWAS) of human populations have identified numerous genetic loci linked with cancer risk and behavior, but nevertheless the major component of cancer heritability remains to be explained. One reason may be that GWAS cannot readily capture gene-gene or gene-environment interactions. Mouse model approaches offer an alternative or complementary strategy, because of our ability to control both the genetic and environmental components of risk. Recently developed genetic tools, including high-throughput technologies such as SNP, CGH and gene expression microarrays, have led to more powerful strategies for refining quantitative trait loci (QTL) and identifying the critical genes. In particular, the cross-species approaches will help to refine locations of QTLs, and reveal their genetic and environmental interactions. The identification of human tumor susceptibility genes and discovery of their roles in carcinogenesis will ultimately be important for the development of methods for prediction of risk, diagnosis, prevention and therapy for human cancers.
Collapse
Affiliation(s)
- Jesús Pérez-Losada
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Instituto Mixto Universidad de Salamanca/CSIC, Campus Miguel de Unamuno s/n, Salamanca, 37007, Spain
| | - Andrés Castellanos-Martín
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Instituto Mixto Universidad de Salamanca/CSIC, Campus Miguel de Unamuno s/n, Salamanca, 37007, Spain
| | - Jian-Hua Mao
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94127, USA
| |
Collapse
|
38
|
Campbell I, Qiu W, Haviv I. Genetic changes in tumour microenvironments. J Pathol 2011; 223:450-8. [PMID: 21294119 DOI: 10.1002/path.2842] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Revised: 12/05/2010] [Accepted: 12/08/2010] [Indexed: 12/21/2022]
Abstract
Numerous in vitro and in vivo studies have established that carcinoma-associated fibroblasts differ phenotypically from fibroblasts associated with normal tissue but the mechanisms underlying these differences are unclear. Since carcinoma-associated fibroblasts can be propagated in vitro for extended periods and still maintain their cancer-promoting phenotype, some investigators have proposed that they might have acquired somatic genetic alterations analogous to those observed in malignant epithelium. Early molecular genetic studies appeared to validate this hypothesis by demonstrating remarkably high frequencies of clonal somatic genetic alterations in carcinoma-associated fibroblasts, including loss of heterozygosity, gene amplification, and point mutations in tumour suppressor genes such as TP53 and PTEN. The initial excitement of these paradigm-changing studies overshadowed concerns that there may have been a more mundane explanation for these observations. In addition to the fact that the data would necessarily invoke an unlikely scenario of the simultaneous generation of two symbiotic malignancies, subsequent molecular genetic studies found no evidence of frequent genomic aberrations. One striking common trait of those studies reporting frequent clonal somatic alterations in carcinoma-associated fibroblasts is the use of tissues and techniques which are well known to be highly prone to generating artefacts such as limiting and poor quality DNA followed by highly multiplexed PCR-based analyses. It is now clear that clonal somatic mutations are not the biological basis of the cancer-promoting attributes of carcinoma-associated fibroblasts.
Collapse
Affiliation(s)
- Ian Campbell
- VBCRC Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
| | | | | |
Collapse
|
39
|
Fibroblast and prostate tumor cell cross-talk: fibroblast differentiation, TGF-β, and extracellular matrix down-regulation. Exp Cell Res 2010; 316:3207-26. [PMID: 20727350 DOI: 10.1016/j.yexcr.2010.08.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Revised: 08/12/2010] [Accepted: 08/12/2010] [Indexed: 01/27/2023]
Abstract
Growth and survival of tumors at a site of metastasis involve interactions with stromal cells in the surrounding environment. Stromal cells aid tumor cell growth by producing cytokines as well as by modifying the environment surrounding the tumor through modulation of the extracellular matrix (ECM). Small leucine-rich proteoglycans (SLRPs) are biologically active components of the ECM which can be altered in the stroma surrounding tumors. The influence tumor cells have on stromal cells has been well elucidated. However, little is understood about the effect metastatic cancer cells have on the cell biology and behavior of the local stromal cells. Our data reveal a significant down-regulation in the expression of ECM components such as collagens I, II, III, and IV, and the SLRPs, decorin, biglycan, lumican, and fibromodulin in stromal cells when grown in the presence of two metastatic prostate cancer cell lines PC3 and DU145. Interestingly, TGF-β down-regulation was observed in stromal cells, as well as actin depolymerization and increased vimentin and α5β1 integrin expression. MT1-MMP expression was upregulated and localized in stromal cell protrusions which extended into the ECM. Moreover, enhanced stromal cell migration was observed after cross-talk with metastatic prostate tumor cells. Xenografting metastatic prostate cancer cells together with "activated" stromal cells led to increased tumorigenicity of the prostate cancer cells. Our findings suggest that metastatic prostate cancer cells create a metastatic niche by altering the phenotype of local stromal cells, leading to changes in the ECM.
Collapse
|
40
|
Welsh M, Moffat L, Jack L, McNeilly A, Brownstein D, Saunders PTK, Sharpe RM, Smith LB. Deletion of androgen receptor in the smooth muscle of the seminal vesicles impairs secretory function and alters its responsiveness to exogenous testosterone and estradiol. Endocrinology 2010; 151:3374-85. [PMID: 20444943 PMCID: PMC3033689 DOI: 10.1210/en.2009-1339] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The seminal vesicles (SVs), like much of the male reproductive tract, depend on androgen-driven stromal-epithelial interactions for normal development, structure, and function. The primary function of the SVs is to synthesize proteins that contribute to the seminal plasma and this is androgen dependent. However, the cell-specific role for androgen action in adult SVs remains unclear. This study analyzed the SV in mice with targeted ablation of androgen receptors specifically in smooth muscle cells (PTM-ARKO) to determine in vivo whether it is androgen action in a subset of the SV stroma, the smooth muscle cells, that drives epithelial function and identity. These mice have significantly smaller SVs in adulthood with less smooth muscle and reduced epithelial cell height. Less epithelial cell proliferation was observed in adult PTM-ARKO SVs, compared with controls, and production of seminal proteins was reduced, indicating global impairment of epithelial cell function in PTM-ARKO SVs. None of these changes could be explained by altered serum testosterone or estradiol concentrations. We also demonstrate altered SV responsiveness to exogenous testosterone and estradiol in PTM-ARKO mice, indicating that smooth muscle androgen receptors may limit the SV epithelial proliferative response to exogenous estrogens. These results therefore demonstrate that the smooth muscle cells play a vital role in androgen-driven stromal-epithelial interactions in the SV, determining epithelial cell structure and function as well as limiting the SV epithelial proliferative response to exogenous estrogens.
Collapse
Affiliation(s)
- Michelle Welsh
- Medical Research Council Human Reproductive Sciences Unit, Centre for Reproductive Biology, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Martin PM, Dussert C, Ouafik L. Stroma : partenaire actif mais sous-estiméde la tumorigenèse, « quand le dialogue remplace le monologue ». ONCOLOGIE 2010. [DOI: 10.1007/s10269-010-1892-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
42
|
Abstract
Tumors are heterogeneous collections of cells with highly variable abilities to survive, grow, and metastasize. This variability likely stems from epigenetic and genetic influences, either stochastic or hardwired by cell type-specific lineage programs. That differentiation underlies tumor cell heterogeneity was elegantly demonstrated in hematopoietic tumors, in which rare primitive cells (cancer stem cells (CSCs)) resembling normal hematopoietic stem cells are ultimately responsible for tumor growth and viability. Because of the compelling clinical implications CSCs pose--across the entire spectrum of cancers--investigators applied the CSC model to cancers arising in tissues with crudely understood differentiation programs. Instead of relying on differentiation, these studies used empirically selected markers and statistical arguments to identify CSCs. The empirical approach has stimulated important questions about "stemness" in cancer cells as well as the validity and stoichiometry of CSC assays. The recent identification of urothelial differentiation programs in urothelial carcinomas (UroCas) supports the idea that solid epithelial cancers (carcinomas) develop and differentiate analogously to normal epithelia and provides new insights about the spatial localization and molecular makeup of carcinoma CSCs. Importantly, CSCs from invasive UroCas (UroCSCs) appear well situated to exchange important signals with adjacent stroma, to escape immune surveillance, and to survive cytotoxic therapy. These signals have potential roles in treatment resistance and many participate in druggable cellular pathways. In this review, we discuss the implications of these findings in understanding CSCs and in better understanding how UroCas form, progress, and should be treated.
Collapse
|
43
|
Yamanouchi H, Umezu T, Tomooka Y. Reconstruction of Oviduct and Demonstration of Epithelial Fate Determination in Mice1. Biol Reprod 2010; 82:528-33. [DOI: 10.1095/biolreprod.109.078329] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
44
|
Follistatin-like-1, a diffusible mesenchymal factor determines the fate of epithelium. Proc Natl Acad Sci U S A 2010; 107:4601-6. [PMID: 20176958 DOI: 10.1073/pnas.0909501107] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mesenchyme is generally believed to play critical roles in "secondary induction" during organogenesis. Because of the complexity of tissue interactions in secondary inductions, however, little is known about the precise mechanisms at the cellular and molecular levels. We have demonstrated that, in mouse oviductal development, the mesenchyme determines the fate of undetermined epithelial cells to become secretory or cilial cells. We have established a model for studying secondary induction by establishing clonal epithelial and mesenchymal cell lines from perinatal p53(-/-) mouse oviducts. The signal sequence trap method collected candidate molecules secreted from mesenchymal cell lines. Naive epithelial cells exposed to Follistatin-like-1 (Fstl1), one of the candidates, became irreversibly committed to expressing a cilial epithelial marker and differentiated into ciliated cells. We concluded that Fstl1 is one of the mesenchymal factors determining oviductal epithelial cell fate. This is a unique demonstration that the determination of epithelial cell fate is induced by a single diffusible factor.
Collapse
|
45
|
Jerde TJ, Bushman W. IL-1 induces IGF-dependent epithelial proliferation in prostate development and reactive hyperplasia. Sci Signal 2009; 2:ra49. [PMID: 19724062 DOI: 10.1126/scisignal.2000338] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Chronic inflammation and reactivation of developmental signaling pathways are both hallmarks of adenocarcinomas. However, developmental and inflammatory processes are generally thought of as distinct and are believed to represent separate paths to carcinogenesis. Here, we show that the inflammatory cytokine interleukin-1alpha (IL-1alpha) plays a critical role in prostate development by activating insulin-like growth factor (IGF) signaling; this process is reiterated during inflammatory reactive hyperplasia to elicit epithelial proliferation. The appearance of developmental signals during hyperplasia supports the hypothesis that reactivation of developmental signaling plays a role in the hyperplasic reaction to inflammation and suggests that there may be a conserved link between inflammatory signaling and canonical developmental pathways.
Collapse
Affiliation(s)
- Travis J Jerde
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA.
| | | |
Collapse
|
46
|
Pottier N, Maurin T, Chevalier B, Puisségur MP, Lebrigand K, Robbe-Sermesant K, Bertero T, Lino Cardenas CL, Courcot E, Rios G, Fourre S, Lo-Guidice JM, Marcet B, Cardinaud B, Barbry P, Mari B. Identification of keratinocyte growth factor as a target of microRNA-155 in lung fibroblasts: implication in epithelial-mesenchymal interactions. PLoS One 2009; 4:e6718. [PMID: 19701459 PMCID: PMC2726943 DOI: 10.1371/journal.pone.0006718] [Citation(s) in RCA: 174] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Accepted: 07/07/2009] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Epithelial-mesenchymal interactions are critical in regulating many aspects of vertebrate embryo development, and for the maintenance of homeostatic equilibrium in adult tissues. The interactions between epithelium and mesenchyme are believed to be mediated by paracrine signals such as cytokines and extracellular matrix components secreted from fibroblasts that affect adjacent epithelia. In this study, we sought to identify the repertoire of microRNAs (miRNAs) in normal lung human fibroblasts and their potential regulation by the cytokines TNF-alpha, IL-1beta and TGF-beta. METHODOLOGY/PRINCIPAL FINDINGS MiR-155 was significantly induced by inflammatory cytokines TNF-alpha and IL-1beta while it was down-regulated by TGF-beta. Ectopic expression of miR-155 in human fibroblasts induced modulation of a large set of genes related to "cell to cell signalling", "cell morphology" and "cellular movement". This was consistent with an induction of caspase-3 activity and with an increase in cell migration in fibroblasts tranfected with miR-155. Using different miRNA bioinformatic target prediction tools, we found a specific enrichment for miR-155 predicted targets among the population of down-regulated transcripts. Among fibroblast-selective targets, one interesting hit was keratinocyte growth factor (KGF, FGF-7), a member of the fibroblast growth factor (FGF) family, which owns two potential binding sites for miR-155 in its 3'-UTR. Luciferase assays experimentally validated that miR-155 can efficiently target KGF 3'-UTR. Site-directed mutagenesis revealed that only one out of the 2 potential sites was truly functional. Functional in vitro assays experimentally validated that miR-155 can efficiently target KGF 3'-UTR. Furthermore, in vivo experiments using a mouse model of lung fibrosis showed that miR-155 expression level was correlated with the degree of lung fibrosis. CONCLUSIONS/SIGNIFICANCE Our results strongly suggest a physiological function of miR-155 in lung fibroblasts. Altogether, this study implicates this miRNA in the regulation by mesenchymal cells of surrounding lung epithelium, making it a potential key player during tissue injury.
Collapse
Affiliation(s)
- Nicolas Pottier
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR6097, Sophia Antipolis, France
- University of Nice Sophia-Antipolis, Nice, France
- EA2679, Faculté de Médecine H. Warembourg, Pôle Recherche, Lille, France
| | - Thomas Maurin
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR6097, Sophia Antipolis, France
- University of Nice Sophia-Antipolis, Nice, France
| | - Benoit Chevalier
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR6097, Sophia Antipolis, France
- University of Nice Sophia-Antipolis, Nice, France
| | - Marie-Pierre Puisségur
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR6097, Sophia Antipolis, France
- University of Nice Sophia-Antipolis, Nice, France
| | - Kevin Lebrigand
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR6097, Sophia Antipolis, France
- University of Nice Sophia-Antipolis, Nice, France
| | - Karine Robbe-Sermesant
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR6097, Sophia Antipolis, France
- University of Nice Sophia-Antipolis, Nice, France
| | - Thomas Bertero
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR6097, Sophia Antipolis, France
- University of Nice Sophia-Antipolis, Nice, France
| | | | - Elisabeth Courcot
- EA2679, Faculté de Médecine H. Warembourg, Pôle Recherche, Lille, France
| | - Géraldine Rios
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR6097, Sophia Antipolis, France
- University of Nice Sophia-Antipolis, Nice, France
| | - Sandra Fourre
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR6097, Sophia Antipolis, France
- University of Nice Sophia-Antipolis, Nice, France
| | | | - Brice Marcet
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR6097, Sophia Antipolis, France
- University of Nice Sophia-Antipolis, Nice, France
| | - Bruno Cardinaud
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR6097, Sophia Antipolis, France
- University of Nice Sophia-Antipolis, Nice, France
| | - Pascal Barbry
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR6097, Sophia Antipolis, France
- University of Nice Sophia-Antipolis, Nice, France
- * E-mail: (PB); (BM)
| | - Bernard Mari
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR6097, Sophia Antipolis, France
- University of Nice Sophia-Antipolis, Nice, France
- * E-mail: (PB); (BM)
| |
Collapse
|
47
|
Zhang H, Li M, Zheng X, Sun Y, Wen Z, Zhao X. Endometriotic stromal cells lose the ability to regulate cell-survival signaling in endometrial epithelial cells in vitro. Mol Hum Reprod 2009; 15:653-63. [PMID: 19684091 DOI: 10.1093/molehr/gap069] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In normal endometrium, stromal factors regulate the growth of epithelial cells. However, epithelial cells in endometriotic lesions display increased proliferation and decreased apoptosis. This work tested the hypothesis that in endometriosis stromal cells lose the ability to regulate survival signaling and cell growth in epithelial cells. Primary normal, endometriotic eutopic and ectopic epithelial cells were cultured in the presence of medium conditioned by normal, eutopic and ectopic endometriotic endometrial stromal cells. Endometriotic epithelial cells showed higher Survivin expression than normal epithelial cells. Conditioned medium (CM) from normal or eutopic endometriotic stromal cells significantly inhibited the Survivin expression and AKt phosphorylation in normal or eutopic endometriotic epithelial cells. However, CM from ectopic endometriotic stromal cells did not have an inhibitory effect on normal or ectopic endometriotic epithelial cells. Inhibition of AKt phosphorylation and Survivin expression in normal or eutopic endometriotic epithelial cells in the presence of stromal factors from normal or eutopic endometriotic stromal cells was enhanced by progesterone, whereas progesterone had little effect in the presence of stromal factors from ectopic endometriotic stromal cells. The inability of ectopic endometriotic stromal cells to regulated PI3K/AKt/Survivin signaling and mediate the progesterone response in endometriotic epithelial cells may facilitate epithelial cell proliferation in endometriosis and promote the survival of endometriotic lesions.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Obstetrics and Gynecology, Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, Shandong 250021, People's Republic of China
| | | | | | | | | | | |
Collapse
|
48
|
Theriot CM, Tove SR, Grunden AM. Biotechnological applications of recombinant microbial prolidases. ADVANCES IN APPLIED MICROBIOLOGY 2009; 68:99-132. [PMID: 19426854 DOI: 10.1016/s0065-2164(09)01203-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Prolidase is a metallopeptidase that is ubiquitous in nature and has been isolated from mammals, bacteria and archaea. Prolidase specifically hydrolyzes dipeptides with a prolyl residue in the carboxy terminus (NH(2)-X-/-Pro-COOH). Currently, the only solved structure of prolidase is from the hyperthermophilic archaeon Pyrococcus furiosus. This enzyme is of particular interest because it can be used in many biotechnological applications. Prolidase is able to degrade toxic organophosphorus (OP) compounds, namely, by cleaving the P-F and P-O bonds in the nerve agents, sarin and soman. Applications using prolidase to detoxify OP nerve agents include its incorporation into fire-fighting foams and as biosensors for OP compound detection. Prolidases are also employed in the cheese-ripening process to improve cheese taste and texture. In humans, prolidase deficiency (PD) is a rare autosomal recessive disorder that affects the connective tissue. Symptoms of PD include skin lesions, mental retardation and recurrent respiratory infections. Enzyme replacement therapies are currently being studied in an effort to optimize enzyme delivery and stability for this application. Previously, prolidase has been linked to collagen metabolism and more recently is being associated with melanoma. Increased prolidase activity in melanoma cell lines has lead investigators to create cancer prodrugs targeting this enzyme. Thus, there are many biotechnological applications using recombinant and native forms of prolidase and this review will describe the biochemical and structural properties of prolidases as well as discuss their most current applications.
Collapse
Affiliation(s)
- Casey M Theriot
- Department of Microbiology, North Carolina State University, Raleigh, North Carolina 27695-7615, USA
| | | | | |
Collapse
|
49
|
Fibbi B, Penna G, Morelli A, Adorini L, Maggi M. Chronic inflammation in the pathogenesis of benign prostatic hyperplasia. ACTA ACUST UNITED AC 2009; 33:475-88. [PMID: 19508330 DOI: 10.1111/j.1365-2605.2009.00972.x] [Citation(s) in RCA: 148] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Benign prostatic hyperplasia (BPH) is a common disorder affecting 50-80% of the aged male population. Androgens and age have been traditionally considered the main determinants of prostate enlargement, but in the last years a potentially important role of chronic inflammation in BPH pathogenesis has emerged. Bacterial and non-infectious chronic prostatitis could represent inciting factors leading to tissue hyperproliferation, possibly via the recently demonstrated antigen-presenting capacity of prostatic stromal cells, enabling them to induce and sustain intraglandular immune responses. The prostate growth-promoting chemokine IL-8 could represent a direct link between chronic prostate inflammation and autocrine/paracrine stromal cell proliferation, in agreement with its marked secretion induced in BPH stromal cells by a combination of Th1 and Th17 cell-derived inflammatory cytokines. BPH stromal cells express the vitamin D receptor (VDR), which is up-regulated by exposure to inflammatory stimuli. The non-hypercalcaemic VDR agonist elocalcitol, shown to arrest BPH development by decreasing the intra-prostatic androgen signalling without directly interfering with systemic androgen action, exerts immunoregulatory and anti-inflammatory properties in different prostatic pathology characterized by growth and inflammation. The mechanism of action of VDR agonists supports an important role of chronic inflammation in BPH pathogenesis and strengthens the concept of these agents as a therapeutic option for pharmacological treatment of BPH.
Collapse
Affiliation(s)
- B Fibbi
- Andrology Unit, Department of Clinical Physiopathology, University of Florence, Florence, Italy
| | | | | | | | | |
Collapse
|
50
|
Arnold JT. DHEA metabolism in prostate: For better or worse? Mol Cell Endocrinol 2009; 301:83-8. [PMID: 19013497 PMCID: PMC2667103 DOI: 10.1016/j.mce.2008.10.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2008] [Revised: 10/14/2008] [Accepted: 10/15/2008] [Indexed: 11/16/2022]
Abstract
Dehydroepiandrosterone (DHEA) is commonly used in the USA as a nutritional supplement for antiaging, metabolic support or other uses. Investigations into understanding the effects of DHEA on human prostate cancer progression have posed more questions than answers and highlight the importance of communications between stromal and epithelial tuoitiuot elements within the prostate that contribute to the regulation of DHEA metabolism. Intracrine metabolism of DHEA to androgens (A) and/or estrogens (E) may occur in one cell compartment (stromal) which may release paracrine hormones or growth/inhibitory factors to the epithelial cells. Alternatively no metabolism of DHEA may occur, resulting in no harmful consequences of high levels of DHEA in prostate tissues. We herein review the tissue components involved and interactions with the prohormone, DHEA and/or resulting metabolites, including dihydrotestosterone (DHT) or 17beta-estradiol (E(2)) in an in vitro model of endocrine-immune-paracrine interactions within the prostate. This work raises questions and hypotheses concerning the role of DHEA in prostate in normal tissues, vs. preneoplastic tissues.
Collapse
Affiliation(s)
- Julia T Arnold
- LCI-Endocrine Section, National Center for Complementary and Alternative Medicine (NCCAM), National Institutes of Health (NIH), Building 10/2B47 MSC 1547, 9000 Rockville Pike, Bethesda, MD 20892-1547, USA.
| |
Collapse
|