1
|
Franchi-Mendes T, Silva M, Carreira MC, Cartaxo AL, Vale P, Karakaidos P, Klinakis A, Fernandes-Platzgummer A, da Silva CL. Xenogeneic-free platform for the isolation and scalable expansion of human bladder smooth muscle cells. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2025; 46:e00878. [PMID: 40094098 PMCID: PMC11909460 DOI: 10.1016/j.btre.2025.e00878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 03/19/2025]
Abstract
Introduction Smooth muscle cells (SMC) play a crucial role in bladder tissue engineering strategies. Scalable, Good Manufacturing Practice (GMP)-compliant platforms are essential for producing clinically relevant cell numbers. Materials & Methods A gamma-irradiated human platelet lysate (HPL) supplement was used to develop a xeno(geneic)-free process for the isolation and scalable expansion of human bladder-derived SMC. Results Cells were isolated using an explant-based technique and expanded ex vivo, expressing typical SMC markers (α-SMA, desmin, caldesmon and SM22-α). Cell culture was successfully scaled-up using spinner flasks combined with plastic microcarriers, starting with a 2.8 × 103 cells/cm2 inoculum (i.e. 1 × 106 cells). Cell-microcarrier adhesion rates exceeded 80% within 24 hours with fold expansion ranging from 3.5 to 16.8 after 7 days, dependent on donor variability. Harvested cells retained their SMC phenotype. Conclusions This xeno-free, GMP compliant process enables scalable manufacturing of human bladder-derived SMC while preserving cell identity, potentially advancing clinical applications in bladder engineering.
Collapse
Affiliation(s)
- Teresa Franchi-Mendes
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Marília Silva
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Maria Catarina Carreira
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Ana Luísa Cartaxo
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Paulo Vale
- Urology Department, Hospital CUF Descobertas, Lisboa, Portugal
| | | | | | - Ana Fernandes-Platzgummer
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia L. da Silva
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
2
|
Lopez TE, Zhang H, Bouysse E, Neiers F, Ye XY, Garrido C, Wendremaire M, Lirussi F. A pivotal role for the IL-1β and the inflammasome in preterm labor. Sci Rep 2024; 14:4234. [PMID: 38378749 PMCID: PMC10879161 DOI: 10.1038/s41598-024-54507-w] [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: 12/12/2023] [Accepted: 02/13/2024] [Indexed: 02/22/2024] Open
Abstract
During labor, monocytes infiltrate massively the myometrium and differentiate into macrophages secreting high levels of reactive oxygen species and of pro-inflammatory cytokines (i.e. IL-1β), leading to myometrial contraction. Although IL-1β is clearly implicated in labor, its function and that of the inflammasome complex that cleaves the cytokine in its active form, has never been studied on steps preceding contraction. In this work, we used our model of lipopolysaccharide-induced preterm labor to highlight their role. We demonstrated that IL-1β was secreted by the human myometrium during labor or in presence of infection and was essential for myometrial efficient contractions as its blockage with an IL-1 receptor antagonist (Anakinra) or a neutralizing antibody completely inhibited the induced contractions. We evaluated the implication of the inflammasome on myometrial contractions and differentiation stages of labor onset. We showed that the effects of macrophage-released IL-1β in myometrial cell transactivation were blocked by inhibition of the inflammasome, suggesting that the inflammasome by producing IL-1β was essential in macrophage/myocyte crosstalk during labor. These findings provide novel innovative approaches in the management of preterm labor, specifically the use of an inflammasome inhibitor to block the precursor stages of labor before the acquisition of the contractile phenotype.
Collapse
Affiliation(s)
- T E Lopez
- INSERM U1231, Labex LIPSTIC and Label of Excellence from la Ligue Nationale Contre le Cancer, 21000, Dijon, France
- Faculty of Medicine and Pharmacy, University of Burgundy, 21000, Dijon, France
| | - H Zhang
- INSERM U1231, Labex LIPSTIC and Label of Excellence from la Ligue Nationale Contre le Cancer, 21000, Dijon, France
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - E Bouysse
- INSERM U1231, Labex LIPSTIC and Label of Excellence from la Ligue Nationale Contre le Cancer, 21000, Dijon, France
- Faculty of Medicine and Pharmacy, University of Burgundy, 21000, Dijon, France
| | - F Neiers
- Faculty of Medicine and Pharmacy, University of Burgundy, 21000, Dijon, France
| | - X Y Ye
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - C Garrido
- INSERM U1231, Labex LIPSTIC and Label of Excellence from la Ligue Nationale Contre le Cancer, 21000, Dijon, France
- Faculty of Medicine and Pharmacy, University of Burgundy, 21000, Dijon, France
- Cancer Center George-François Leclerc, 21000, Dijon, France
| | - M Wendremaire
- INSERM U1231, Labex LIPSTIC and Label of Excellence from la Ligue Nationale Contre le Cancer, 21000, Dijon, France
- Faculty of Medicine and Pharmacy, University of Burgundy, 21000, Dijon, France
| | - Frédéric Lirussi
- INSERM U1231, Labex LIPSTIC and Label of Excellence from la Ligue Nationale Contre le Cancer, 21000, Dijon, France.
- Laboratory of Pharmacology-Toxicology, Platform PACE, University Hospital Besançon, 25000, Besançon, France.
- Faculty of Medicine and Pharmacy, University of Franche-Comté, 25000, Besançon, France.
| |
Collapse
|
3
|
Boros-Rausch A, Shynlova O, Lye SJ. A Broad-Spectrum Chemokine Inhibitor Blocks Inflammation-Induced Myometrial Myocyte-Macrophage Crosstalk and Myometrial Contraction. Cells 2021; 11:cells11010128. [PMID: 35011690 PMCID: PMC8750067 DOI: 10.3390/cells11010128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 01/10/2023] Open
Abstract
Prophylactic administration of the broad-spectrum chemokine inhibitor (BSCI) FX125L has been shown to suppress uterine contraction, prevent preterm birth (PTB) induced by Group B Streptococcus in nonhuman primates, and inhibit uterine cytokine/chemokine expression in a murine model of bacterial endotoxin (LPS)-induced PTB. This study aimed to determine the mechanism(s) of BSCI action on human myometrial smooth muscle cells. We hypothesized that BSCI prevents infection-induced contraction of uterine myocytes by inhibiting the secretion of pro-inflammatory cytokines, the expression of contraction-associated proteins and disruption of myocyte interaction with tissue macrophages. Myometrial biopsies and peripheral blood were collected from women at term (not in labour) undergoing an elective caesarean section. Myocytes were isolated and treated with LPS with/out BSCI; conditioned media was collected; cytokine secretion was analyzed by ELISA; and protein expression was detected by immunoblotting and immunocytochemistry. Functional gap junction formation was assessed by parachute assay. Collagen lattices were used to examine myocyte contraction with/out blood-derived macrophages and BSCI. We found that BSCI inhibited (1) LPS-induced activation of transcription factor NF-kB; (2) secretion of chemokines (MCP-1/CCL2 and IL-8/CXCL8); (3) Connexin43-mediated intercellular connectivity, thereby preventing myocyte–macrophage crosstalk; and (4) myocyte contraction. BSCI represents novel therapeutics for prevention of inflammation-induced PTB in women.
Collapse
Affiliation(s)
- Adam Boros-Rausch
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, 25 Orde Street, Suite 6-1017, Toronto, ON M5G 1X5, Canada; (A.B.-R.); (S.J.L.)
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Oksana Shynlova
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, 25 Orde Street, Suite 6-1017, Toronto, ON M5G 1X5, Canada; (A.B.-R.); (S.J.L.)
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Obstetrics & Gynecology, University of Toronto, Toronto, ON M5S 1A1, Canada
- Correspondence: ; Tel.: +1-416-586-4800 (ext. 5635); Fax: +1-416-586-5116
| | - Stephen James Lye
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, 25 Orde Street, Suite 6-1017, Toronto, ON M5G 1X5, Canada; (A.B.-R.); (S.J.L.)
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Obstetrics & Gynecology, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
4
|
Wendremaire M, Hadi T, Pezze M, Barrichon M, Lopez T, Neiers F, Sagot P, Garrido C, Lirussi F. Macrophage-induced reactive oxygen species promote myometrial contraction and labor-associated mechanisms†. Biol Reprod 2021; 102:1326-1339. [PMID: 32167534 DOI: 10.1093/biolre/ioaa032] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 12/31/2019] [Accepted: 03/12/2020] [Indexed: 11/14/2022] Open
Abstract
At labor, the myometrium is infiltrated by a massive influx of macrophages that secrete high levels of pro-inflammatory cytokines inducing the expression of specific labor-associated markers. However, the interactions between myocytes and macrophages and the role of macrophages in the myometrium at labor remain to be elucidated. In this work, we studied the role of myometrium-infiltrated macrophages and their interaction with myocytes in lipopolysaccharide-induced preterm labor. A co-culture model of human primary myometrial cells and macrophages was developed and validated. Collagen lattices were used to evaluate myocyte contraction. Differentiation steps were assessed by (i) phalloidin and vinculin staining for cytoskeleton reorganization, (ii) gap junction protein alpha 1 expression and scrape loading/dye transfer with Lucifer Yellow for gap junction intercellular communication, and (iii) calcium imaging for cell excitability. We demonstrated that macrophages favored lipopolysaccharide-induced contraction and early differentiation of myometrial cells. Transwell assays showed that previous activation of macrophages by lipopolysaccharide was essential for this differentiation and that macrophage/myocyte interactions involved macrophage release of reactive oxygen species (ROS). The effects of macrophage-released ROS in myometrial cell transactivation were mimicked by H2O2, suggesting that superoxide anion is a major intermediate messenger in macrophage/myocyte crosstalk during labor. These novel findings provide the foundation for innovative approaches to managing preterm labor, specifically the use of antioxidants to inhibit the initial stages of labor before the contractile phenotype has been acquired. In addition, the co-culture model developed by our team could be used in future research to decipher pathophysiological signaling pathways or screen/develop new tocolytics.
Collapse
Affiliation(s)
- Maeva Wendremaire
- Institut National de la Santéc et de la Recherche Médicale, Lipides Nutrition Cancer, Dijon, France.,Université de Bourgogne Franche-Comté, Lipides Nutrition Cancer, Dijon, France.,Laboratoire de Pharmacologie-Toxicologie, CHU Dijon-Bourgogne, Dijon, France
| | - Tarik Hadi
- Institut National de la Santéc et de la Recherche Médicale, Lipides Nutrition Cancer, Dijon, France.,Université de Bourgogne Franche-Comté, Lipides Nutrition Cancer, Dijon, France.,Department of Cardiac Surgery, NYU Langone Medical Center, New York, NY, USA
| | - Maria Pezze
- Institut National de la Santéc et de la Recherche Médicale, Lipides Nutrition Cancer, Dijon, France.,Université de Bourgogne Franche-Comté, Lipides Nutrition Cancer, Dijon, France
| | - Marina Barrichon
- Institut National de la Santéc et de la Recherche Médicale, Lipides Nutrition Cancer, Dijon, France.,Université de Bourgogne Franche-Comté, Lipides Nutrition Cancer, Dijon, France
| | - Tatiana Lopez
- Institut National de la Santéc et de la Recherche Médicale, Lipides Nutrition Cancer, Dijon, France.,Université de Bourgogne Franche-Comté, Lipides Nutrition Cancer, Dijon, France
| | - Fabrice Neiers
- Centre des Sciences du Goût et de l'Alimentation, INRA, CNRS, Université de Bourgogne Franche-Comté, Dijon, France
| | - Paul Sagot
- Service de Gynécologie-Obstétrique, CHU Dijon-Bourgogne, Dijon, France
| | - Carmen Garrido
- Institut National de la Santéc et de la Recherche Médicale, Lipides Nutrition Cancer, Dijon, France.,Université de Bourgogne Franche-Comté, Lipides Nutrition Cancer, Dijon, France.,Centre Georges François Leclerc, Dijon, France
| | - Frédéric Lirussi
- Institut National de la Santéc et de la Recherche Médicale, Lipides Nutrition Cancer, Dijon, France.,Université de Bourgogne Franche-Comté, Lipides Nutrition Cancer, Dijon, France.,Laboratoire de Pharmacologie-Toxicologie, CHU Dijon-Bourgogne, Dijon, France
| |
Collapse
|
5
|
Zierden HC, Ortiz JI, DeLong K, Yu J, Li G, Dimitrion P, Bensouda S, Laney V, Bailey A, Anders NM, Scardina M, Mahendroo M, Mesiano S, Burd I, Wagner G, Hanes J, Ensign LM. Enhanced drug delivery to the reproductive tract using nanomedicine reveals therapeutic options for prevention of preterm birth. Sci Transl Med 2021; 13:13/576/eabc6245. [PMID: 33441428 DOI: 10.1126/scitranslmed.abc6245] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022]
Abstract
Inflammation contributes to nearly 4 million global premature births annually. Here, we used a mouse model of intrauterine inflammation to test clinically used formulations, as well as engineered nanoformulations, for the prevention of preterm birth (PTB). We observed that neither systemic 17a-hydroxyprogesterone caproate (Makena) nor vaginal progesterone gel (Crinone) was sufficient to prevent inflammation-induced PTB, consistent with recent clinical trial failures. However, we found that vaginal delivery of mucoinert nanosuspensions of histone deacetylase (HDAC) inhibitors, in some cases with the addition of progesterone, prevented PTB and resulted in delivery of live pups exhibiting neurotypical development. In human myometrial cells in vitro, the P4/HDAC inhibitor combination both inhibited cell contractility and promoted the anti-inflammatory action of P4 by increasing progesterone receptor B stability. Here, we demonstrate the use of vaginally delivered drugs to prevent intrauterine inflammation-induced PTB resulting in the birth of live offspring in a preclinical animal model.
Collapse
Affiliation(s)
- Hannah C Zierden
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jairo I Ortiz
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.,Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Kevin DeLong
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.,Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jingqi Yu
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Gaoshan Li
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Peter Dimitrion
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Sabrine Bensouda
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Victoria Laney
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Anna Bailey
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Nicole M Anders
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Morgan Scardina
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mala Mahendroo
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sam Mesiano
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Gunter Wagner
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06520, USA
| | - Justin Hanes
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.,Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Laura M Ensign
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA. .,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.,Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD 21287, USA
| |
Collapse
|
6
|
Progesterone receptor isoform B regulates the Oxtr- Plcl2- Trpc3 pathway to suppress uterine contractility. Proc Natl Acad Sci U S A 2021; 118:2011643118. [PMID: 33707208 DOI: 10.1073/pnas.2011643118] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Uterine contractile dysfunction leads to pregnancy complications such as preterm birth and labor dystocia. In humans, it is hypothesized that progesterone receptor isoform PGR-B promotes a relaxed state of the myometrium, and PGR-A facilitates uterine contraction. This hypothesis was tested in vivo using transgenic mouse models that overexpress PGR-A or PGR-B in smooth muscle cells. Elevated PGR-B abundance results in a marked increase in gestational length compared to control mice (21.1 versus 19.1 d respectively, P < 0.05). In both ex vivo and in vivo experiments, PGR-B overexpression leads to prolonged labor, a significant decrease in uterine contractility, and a high incidence of labor dystocia. Conversely, PGR-A overexpression leads to an increase in uterine contractility without a change in gestational length. Uterine RNA sequencing at midpregnancy identified 1,174 isoform-specific downstream targets and 424 genes that are commonly regulated by both PGR isoforms. Gene signature analyses further reveal PGR-B for muscle relaxation and PGR-A being proinflammatory. Elevated PGR-B abundance reduces Oxtr and Trpc3 and increases Plcl2 expression, which manifests a genetic profile of compromised oxytocin signaling. Functionally, both endogenous PLCL2 and its paralog PLCL1 can attenuate uterine muscle cell contraction in a CRISPRa-based assay system. These findings provide in vivo support that PGR isoform levels determine distinct transcriptomic landscapes and pathways in myometrial function and labor, which may help further the understanding of abnormal uterine function in the clinical setting.
Collapse
|
7
|
Maxey AP, McCain ML. Tools, techniques, and future opportunities for characterizing the mechanobiology of uterine myometrium. Exp Biol Med (Maywood) 2021; 246:1025-1035. [PMID: 33554648 DOI: 10.1177/1535370221989259] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The myometrium is the smooth muscle layer of the uterus that generates the contractions that drive processes such as menstruation and childbirth. Aberrant contractions of the myometrium can result in preterm birth, insufficient progression of labor, or other difficulties that can lead to maternal or fetal complications or even death. To investigate the underlying mechanisms of these conditions, the most common model systems have conventionally been animal models and human tissue strips, which have limitations mostly related to relevance and scalability, respectively. Myometrial smooth muscle cells have also been isolated from patient biopsies and cultured in vitro as a more controlled experimental system. However, in vitro approaches have focused primarily on measuring the effects of biochemical stimuli and neglected biomechanical stimuli, despite the extensive evidence indicating that remodeling of tissue rigidity or excessive strain is associated with uterine disorders. In this review, we first describe the existing approaches for modeling human myometrium with animal models and human tissue strips and compare their advantages and disadvantages. Next, we introduce existing in vitro techniques and assays for assessing contractility and summarize their applications in elucidating the role of biochemical or biomechanical stimuli on human myometrium. Finally, we conclude by proposing the translation of "organ on chip" approaches to myometrial smooth muscle cells as new paradigms for establishing their fundamental mechanobiology and to serve as next-generation platforms for drug development.
Collapse
Affiliation(s)
- Antonina P Maxey
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
8
|
Gnecco JS, Brown AT, Kan EL, Baugh L, Ives C, Loring M, Griffith LG. Physiomimetic Models of Adenomyosis. Semin Reprod Med 2020; 38:179-196. [PMID: 33176387 PMCID: PMC7803459 DOI: 10.1055/s-0040-1719084] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adenomyosis remains an enigmatic disease in the clinical and research communities. The high prevalence, diversity of morphological and symptomatic presentations, array of potential etiological explanations, and variable response to existing interventions suggest that different subgroups of patients with distinguishable mechanistic drivers of disease may exist. These factors, combined with the weak links to genetic predisposition, make the entire spectrum of the human condition challenging to model in animals. Here, after an overview of current approaches, a vision for applying physiomimetic modeling to adenomyosis is presented. Physiomimetics combines a system's biology analysis of patient populations to generate hypotheses about mechanistic bases for stratification with in vitro patient avatars to test these hypotheses. A substantial foundation for three-dimensional (3D) tissue engineering of adenomyosis lesions exists in several disparate areas: epithelial organoid technology; synthetic biomaterials matrices for epithelial–stromal coculture; smooth muscle 3D tissue engineering; and microvascular tissue engineering. These approaches can potentially be combined with microfluidic platform technologies to model the lesion microenvironment and can potentially be coupled to other microorgan systems to examine systemic effects. In vitro patient-derived models are constructed to answer specific questions leading to target identification and validation in a manner that informs preclinical research and ultimately clinical trial design.
Collapse
Affiliation(s)
- Juan S Gnecco
- Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Alex T Brown
- Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Ellen L Kan
- Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Lauren Baugh
- Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Clara Ives
- Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Megan Loring
- Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Endometriosis and Adenomyosis Care Collaborative, Center for Minimally Invasive Gynecologic Surgery, Newton Wellesley Hospital, Newton, Massachusetts
| | - Linda G Griffith
- Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
9
|
Transplantation of hPSC-derived pericyte-like cells promotes functional recovery in ischemic stroke mice. Nat Commun 2020; 11:5196. [PMID: 33060592 PMCID: PMC7566513 DOI: 10.1038/s41467-020-19042-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 09/26/2020] [Indexed: 12/15/2022] Open
Abstract
Pericytes play essential roles in blood–brain barrier (BBB) integrity and dysfunction or degeneration of pericytes is implicated in a set of neurological disorders although the underlying mechanism remains largely unknown. However, the scarcity of material sources hinders the application of BBB models in vitro for pathophysiological studies. Additionally, whether pericytes can be used to treat neurological disorders remains to be elucidated. Here, we generate pericyte-like cells (PCs) from human pluripotent stem cells (hPSCs) through the intermediate stage of the cranial neural crest (CNC) and reveal that the cranial neural crest-derived pericyte-like cells (hPSC-CNC PCs) express typical pericyte markers including PDGFRβ, CD146, NG2, CD13, Caldesmon, and Vimentin, and display distinct contractile properties, vasculogenic potential and endothelial barrier function. More importantly, when transplanted into a murine model of transient middle cerebral artery occlusion (tMCAO) with BBB disruption, hPSC-CNC PCs efficiently promote neurological functional recovery in tMCAO mice by reconstructing the BBB integrity and preventing of neuronal apoptosis. Our results indicate that hPSC-CNC PCs may represent an ideal cell source for the treatment of BBB dysfunction-related disorders and help to model the human BBB in vitro for the study of the pathogenesis of such neurological diseases. Pericytes play an essential role in blood brain barrier (BBB) integrity. Here, the authors generate pericyte-like cells (PCs) from human pluripotent stem cells (hPSCs) which display functional properties and also promote BBB recovery in a mouse model of cerebral artery occlusion.
Collapse
|
10
|
Modzelewska B, Jóźwik M, Jóźwik M, Tylicka M, Kleszczewski T. The effects of extended nitric oxide release on responses of the human non-pregnant myometrium to endothelin-1 or vasopressin. Pharmacol Rep 2019; 71:892-898. [PMID: 31419630 DOI: 10.1016/j.pharep.2019.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 04/12/2019] [Accepted: 05/06/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND Uterotonic mediators: endothelin-1 (ET-1), arginine vasopressin (AVP), and nitric oxide (NO) play important roles in the regulation of uterine contractility. We hypothesize that NO affects both ET-1 or AVP. Therefore, this study investigated the involvement of extended exogenous NO release in the regulation of responses of the human non-pregnant myometrium to ET-1 and AVP. METHODS Specimens were obtained from 10 premenopausal women, undergoing hysterectomy for benign gynecological disorders. Responses of the myometrial strips to ET-1 or AVP in the absence and presence of an exogenous NO donor (diethylenetriamine; DETA/NO; 10-4 mol/L) were recorded under isometric conditions. To inhibit endogenous NO, a competitive inhibitor of NO synthase, L-NG-nitroarginine (L-NNA) was added to the organ bath. RESULTS ET-1 enhanced the spontaneous contractile activity of the myometrium more powerfully (p < 0.01) than AVP. Preincubation with exogenous NO weakened ET-1- or AVP-induced increases in this contractile activity (p < 0.05). However, unexpected results were obtained after preincubation with L-NNA and with DETA/NO then added. Both ET-1 and AVP induced augmented contractile effects in almost all concentrations compared with the responses to these peptides alone or after NOS synthase inhibition (both p < 0.01). CONCLUSIONS This study demonstrated for the first time that extended incubation with a NO donor influences the uterine muscle response evoked by ET-1 and AVP. Both endogenous and exogenous NO is involved in the control of the uterine responses to ET-1 or AVP of non-pregnant myometrium. Furthermore, both peptides stimulate increased uterine contractility when the local imbalance between the constrictive and relaxing mediators takes place.
Collapse
Affiliation(s)
- Beata Modzelewska
- Department of Biophysics, Medical University of Białystok, Białystok, Poland.
| | - Maciej Jóźwik
- Department of Gynecology and Gynecologic Oncology, Medical University of Białystok, Białystok, Poland
| | - Marcin Jóźwik
- Department of Gynecology and Obstetrics, Faculty of Medical Sciences, University of Warmia and Mazury, Olsztyn, Poland
| | - Marzena Tylicka
- Department of Biophysics, Medical University of Białystok, Białystok, Poland
| | - Tomasz Kleszczewski
- Department of Biophysics, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
11
|
Drew T, Balki M. What does basic science tell us about the use of uterotonics? Best Pract Res Clin Obstet Gynaecol 2019; 61:3-14. [PMID: 31326333 DOI: 10.1016/j.bpobgyn.2019.05.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 05/25/2019] [Accepted: 05/27/2019] [Indexed: 01/05/2023]
Abstract
Pharmacotherapy with uterotonics remains the mainstay of the management for post-partum haemorrhage. Clinical studies evaluating the efficacy of these drugs are fraught with confounders, which may influence uterine contractility and blood loss. For this reason, a range of techniques have been developed to study myometrial function in vitro, allowing for the comparison of various drugs in a controlled-simulated physiological environment. In this review, we focus on the main classes of uterotonic drugs and outline their molecular and physiological basis of action. We explore the evidence related to appropriate drug dosing and relative efficacy, and compare the evidence gleaned from clinical and in vitro studies. We discuss the mechanism of oxytocin desensitisation and how basic science has helped us understand this phenomenon. We also discuss the in vitro research findings for each of the main classes of uterotonic drugs that have contributed to an improved understanding of the management of post-partum haemorrhage and, ultimately, better care for mothers.
Collapse
Affiliation(s)
- Thomas Drew
- Department of Anesthesia and Pain Management, Mount Sinai Hospital, University of Toronto, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada.
| | - Mrinalini Balki
- Department of Anesthesia and Pain Management, Department of Obstetrics and Gynaecology, University of Toronto, The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada.
| |
Collapse
|
12
|
Mihanfar A, Sadigh AR, Fattahi A, Latifi Z, Hasanzadeh-Moghadam M, Samadi M, Farzadi L, Hamdi K, Ghasemzadeh A, Nejabati HR, Nouri M. Endothelins and their receptors in embryo implantation. J Cell Biochem 2019; 120:14274-14284. [PMID: 31106465 DOI: 10.1002/jcb.28983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 03/31/2019] [Accepted: 04/08/2019] [Indexed: 12/12/2022]
Abstract
As a critical stage of pregnancy, the implantation of blastocysts into the endometrium is a progressive, excessively regulated local tissue remodeling step involving a complex sequence of genetic and cellular interplay executed within an optimal time frame. For better understanding the causes of infertility and, more importantly, for developing powerful strategies for successful implantations and combating infertility, an increasing number of recent studies have been focused on the identification and study of newly described substances in the reproductive tree. The endothelins (ET), a 21-aminoacidic family of genes, have been reported to be responsible for the contraction of vascular and nonvascular smooth muscles, including the smooth muscles of the uterus. Therefore, this review aims to comprehensively discuss the physiological role of endothelins and signaling through their receptors, as well as their probable involvement in the implantation process.
Collapse
Affiliation(s)
- Aynaz Mihanfar
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Aydin Raei Sadigh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Latifi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mahrokh Samadi
- Nephrology and Kidney Transplant Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Laya Farzadi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kobra Hamdi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aliyeh Ghasemzadeh
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Reza Nejabati
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
13
|
Heidari Kani M, Chan EC, Young RC, Butler T, Smith R, Paul JW. 3D Cell Culturing and Possibilities for Myometrial Tissue Engineering. Ann Biomed Eng 2016; 45:1746-1757. [PMID: 27770218 DOI: 10.1007/s10439-016-1749-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 10/12/2016] [Indexed: 12/19/2022]
Abstract
Research insights into uterine function and the mechanisms of labour have been hindered by the lack of suitable animal and cellular models. The use of traditional culturing methods limits the exploration of complex uterine functions, such as cell interactions, connectivity and contractile behaviour, as it fails to mimic the three-dimensional (3D) nature of uterine cell interactions in vivo. Animal models are an option, however, use of these models is constrained by ethical considerations as well as translational limitations to humans. Evidence indicates that these limitations can be overcome by using 3D culture systems, or 3D Bioprinters, to model the in vivo cytological architecture of the tissue in an in vitro environment. 3D cultured or 3D printed cells can be used to form an artificial tissue. This artificial tissue can not only be used as an appropriate model in which to study cellular function and organisation, but could also be used for regenerative medicine purposes including organ or tissue transplantation, organ donation and obstetric care. The current review describes recent developments in cell culture that can facilitate the development of myometrial 3D structures and tissue engineering applications.
Collapse
Affiliation(s)
- Minoo Heidari Kani
- Mothers and Babies Research Centre, School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia. .,Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia. .,Priority Research Centre of Reproductive Science, University of Newcastle, Callaghan, NSW, 2308, Australia.
| | - Eng-Cheng Chan
- Mothers and Babies Research Centre, School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia.,Priority Research Centre of Reproductive Science, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Roger C Young
- Mothers and Babies Research Centre, School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Trent Butler
- Mothers and Babies Research Centre, School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia.,Priority Research Centre of Reproductive Science, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Roger Smith
- Mothers and Babies Research Centre, School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia.,Priority Research Centre of Reproductive Science, University of Newcastle, Callaghan, NSW, 2308, Australia.,John Hunter Hospital, New Lambton Heights, NSW, 2305, Australia
| | - Jonathan W Paul
- Mothers and Babies Research Centre, School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia.,Priority Research Centre of Reproductive Science, University of Newcastle, Callaghan, NSW, 2308, Australia
| |
Collapse
|
14
|
ZHANG YANXIA, GUO JUFANG, LIN CHUAN, LU LU, LI CHENGZHI. Effect of low-intensity focused ultrasound on endothelin-1, nitrogen monoxide and oxytocin receptor in the uterine tissues of Sprague-Dawley rats following abortion. Biomed Rep 2016; 4:340-344. [DOI: 10.3892/br.2016.581] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 11/11/2015] [Indexed: 12/14/2022] Open
|
15
|
Voltolini C, Battersby S, Novembri R, Torricelli M, Severi FM, Petraglia F, Norman JE. Urocortin 2 role in placental and myometrial inflammatory mechanisms at parturition. Endocrinology 2015; 156:670-9. [PMID: 25426872 DOI: 10.1210/en.2014-1432] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The purpose of the study was to investigate urocortin (Ucn)2 involvement in placental and myometrial inflammatory pathways associated with parturition by evaluating: 1) Ucn2 and its receptor, CRH-receptor type 2 (CRH-R2), expression in laboring/nonlaboring human gestational tissues and in mouse utero-placental tissues approaching delivery; and 2) Ucn2 effect on myometrial contractility and on the expression of inflammatory mediators (prostaglandin F2α receptor and cytokines) and regulation of Ucn2 by TNF-α in cultured myometrial cell line. Placenta (n = 16), fetal membranes (n = 16), and myometrium (n = 22) were obtained from healthy pregnant women delivering at term by vaginal/elective caesarean delivery and from timed-pregnant mice on days 16-19. Expression of Ucn2/CRH-R2 in human/mouse tissues and inflammatory mediators in myometrial cell lines were measured by RT-PCR or ELISA, mouse Ucn2/CRH-R2 protein localization by immunohistochemistry. Ucn2 but not CRH-R2 was up-regulated (P < .05) in all human tissues in labor (compared with before labor) and increased significantly (P < .01) in mouse placenta approaching delivery. Ucn2 was up-regulated by TNF-α via nuclear factor-κB (NF-kB) in myometrium cell lines (P < .05 or P < .01 on the basis of treatment doses) and increased proinflammatory mediators and prostaglandin F (PGF2α) receptor expression (P < .05) via CRH-R2, without a direct effect on contractility. Placental and myometrial Ucn2 may play a role in the endocrine-inflammatory processes of parturition, representing a potential target for treating inflammation-induced obstetric complications.
Collapse
Affiliation(s)
- Chiara Voltolini
- Department of Molecular and Developmental Medicine (C.V., R.N., M.T., F.M.S., F.P.), University of Siena, 53100 Siena, Italy; and Medical Research Council Centre for Reproductive Health (S.B., J.E.N.), University of Edinburgh, EH16 4TY Edinburgh, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
16
|
Adverse effects of 4-tert-octylphenol on the production of oxytocin and hCG in pregnant rats. Lab Anim Res 2014; 30:123-30. [PMID: 25324873 PMCID: PMC4188831 DOI: 10.5625/lar.2014.30.3.123] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 09/03/2014] [Accepted: 09/04/2014] [Indexed: 01/31/2023] Open
Abstract
Endocrine-disrupting chemicals (EDCs) are exogenous substances that alter the structure or function of the endocrine system. 4-Tert-octylphenol (OP) is one of the most representative EDCs and has estrogenic effects. In this study, we examined the effects of ethinyl estradiol (EE) and OP on the pituitary gland, placenta, and uterus of pregnant rats. Expression levels of human chorionic gonadotropin (hCG), oxytocin (OT), and contraction-associated proteins (CAPs) were determined, and uterine contractile activity was measured by uterine contraction assay. EE and OP both increased mRNA expression of OT and hCG in the pituitary gland but not the placenta. Since OT and hCG control uterine contraction, we next examined CAP expression in the uterus. Expression of 15-hydroxyprostaglandin-dehydrogenase (PGDH) was upregulated by OP, whereas expression of other CAPs was unaffected. To clarify the effect of OP on uterine contraction in pregnant rats, uterine contraction assay was performed. The 17β-Estradiol (E2) did not affect contraction of primary uterine cells harvested from pregnant rats in a 3D collagen gel model. However, OP showed different effects from E2 by significantly reducing contraction activity. In summary, we demonstrated that OP interferes with regulation of OT and hCG in the pituitary gland as well as PGDH in the uterus, thereby reducing uterine contraction activity. This result differs from the action of endogenous E2. Collectively, these findings suggest that exposure to EDCs such as OP during pregnancycan reduce uterine contractile ability, which may result in contraction-associated adverse effects such as metratonia, bradytocia, and uterine leiomyomata.
Collapse
|
17
|
Vink JY, Charles-Horvath PC, Kitajewski JK, Reeves CV. Anthrax toxin receptor 2 promotes human uterine smooth muscle cell viability, migration and contractility. Am J Obstet Gynecol 2014; 210:154.e1-8. [PMID: 24060446 DOI: 10.1016/j.ajog.2013.09.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Revised: 08/21/2013] [Accepted: 09/19/2013] [Indexed: 01/26/2023]
Abstract
OBJECTIVE Previously we demonstrated anthrax toxin receptor 2 knockout (Antxr2(-/-)) mice are fertile but fail to deliver their pups at term. This parturition defect is associated with overaccumulation of extracellular matrix proteins and decreased myometrial cell content in the uterus. Myometrial cell loss in Antxr2(-/-) uterine tissue prompted us to evaluate if ANTXR2 is essential for human uterine smooth muscle cell viability and function. STUDY DESIGN We subjected human uterine smooth muscle cell to lentiviral-mediated knock down or retroviral-mediated overexpression of ANTXR2. Flow cytometry confirmed lentiviral-mediated knock down or retroviral-mediated overexpression in cell lines vs control. Cell behavior and function in control, lentiviral-mediated knock down and retroviral-mediated overexpression cells were evaluated for apoptosis via TUNEL assay, migration via Boyden chamber assay and with oxytocin-mediated collagen contraction assays. Matrix metalloproteinase activity was evaluated using gelatin zymography. Cell lines and samples were run in duplicate. Student t test was used for statistical analysis. RESULTS ANTXR2 is expressed by human uterine smooth muscle cell. Human uterine smooth muscle cell-lentiviral-mediated knock down cells exhibited increased apoptosis (P < .05) and decreased migration (P < .05), although human uterine smooth muscle cell-retroviral-mediated overexpression cells exhibited no change in apoptosis (P = .91) and increased migration (P = .05) vs control. Human uterine smooth muscle cell-lentiviral-mediated knock down cells contracted significantly less than control, although human uterine smooth muscle cell-retroviral-mediated overexpression cells showed no difference in contractility vs control. Matrix metalloproteinase activity 2 activity appeared slightly decreased in human uterine smooth muscle cell-lentiviral-mediated knock down cells and increased in human uterine smooth muscle cell-retroviral-mediated overexpression cells vs control. CONCLUSION ANTXR2 is expressed by human uterine smooth muscle cell and appears important for normal human uterine smooth muscle cell viability, migration and contractility. Further studies are needed to delineate if ANTXR2 is important for normal and abnormal labor patterns.
Collapse
|
18
|
Dismuke WM, Liang J, Overby DR, Stamer WD. Concentration-related effects of nitric oxide and endothelin-1 on human trabecular meshwork cell contractility. Exp Eye Res 2013; 120:28-35. [PMID: 24374036 DOI: 10.1016/j.exer.2013.12.012] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 12/12/2013] [Accepted: 12/16/2013] [Indexed: 12/21/2022]
Abstract
The contractility status of trabecular meshwork (TM) cells influences aqueous humor outflow resistance and intraocular pressure. Using human TM cells as a model, the goal of the present study was to examine concentration-response relationships of two prototypical molecules, nitric oxide (NO) and endothelin-1 (ET-1), known to differentially influence vascular smooth muscle contractility. Efficacy of ET-1, two NO donors (DETA-NO and SNP) and a cGMP analog (8-Br-cGMP) were assessed using two complementary methods: functionally in a gel contraction assay and biochemically using a myosin light chain phosphorylation assay. The NO donors DETA-NO and SNP dose dependently relaxed cultured human TM cells (EC50 for DETA-NO = 6.0 ± 2.4 μM, SNP = 12.6 ± 8.8 μM), with maximum effects at 100 μM. Interestingly, at concentrations of NO donors above 100 μM, the relaxing effect was lost. Relaxation caused by DETA-NO (100 μM) was dose dependently blocked by the soluble guanylate cyclase specific inhibitor ODQ (IC50 = 460 ± 190 nM). In contrast to the NO donors, treatment of cells with the cGMP analog, 8-Br-cGMP produced the largest relaxation (109.4%) that persisted at high concentrations (EC50 = 110 ± 40 μM). ET-1 caused a dose-dependent contraction of human TM cells (EC50 = 1.5 ± 0.5 pM), with maximum effect at 100 pM (56.1%) and this contraction was reversed by DETA-NO (100 μM). Consistent with functional data, phosphorylation status of myosin light chain was dose dependently reduced with DETA-NO, and increased with ET-1. Together, data show that TM cells rapidly change their contractility status over a wide dynamic range, well suited for the regulation of outflow resistance and intraocular pressure.
Collapse
Affiliation(s)
| | - Jin Liang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Darryl R Overby
- Department of Bioengineering, Imperial College London, London, UK
| | - W Daniel Stamer
- Department of Ophthalmology, Duke University, Durham, NC, USA; Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
19
|
Hutchinson JL, Rajagopal SP, Yuan M, Norman JE. Lipopolysaccharide promotes contraction of uterine myocytes
via
activation of Rho/ROCK signaling pathways. FASEB J 2013; 28:94-105. [DOI: 10.1096/fj.13-237040] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- James L. Hutchinson
- Medical Research Council Centre for Reproductive HealthUniversity of EdinburghThe Queen's Medical Research InstituteEdinburghUK
| | - Shalini P. Rajagopal
- Medical Research Council Centre for Reproductive HealthUniversity of EdinburghThe Queen's Medical Research InstituteEdinburghUK
| | - Mei Yuan
- Medical Research Council Centre for Reproductive HealthUniversity of EdinburghThe Queen's Medical Research InstituteEdinburghUK
| | - Jane E. Norman
- Medical Research Council Centre for Reproductive HealthUniversity of EdinburghThe Queen's Medical Research InstituteEdinburghUK
| |
Collapse
|
20
|
An BS, Ahn HJ, Kang HS, Jung EM, Yang H, Hong EJ, Jeung EB. Effects of estrogen and estrogenic compounds, 4-tert-octylphenol, and bisphenol A on the uterine contraction and contraction-associated proteins in rats. Mol Cell Endocrinol 2013; 375:27-34. [PMID: 23664861 DOI: 10.1016/j.mce.2013.04.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 04/09/2013] [Accepted: 04/26/2013] [Indexed: 12/13/2022]
Abstract
We examined the effects of estradiol (E2), 4-tert-octylphenol (OP), and bisphenol A (BPA) on uterine contractions in immature rats. The expression and localization of contraction-associated proteins (CAPs), and contractility of rat uterus with a collagen gel contraction assay were analyzed. E2, OP, and BPA all increased oxytocin (OT)-related pathway, while the prostaglandin-related signaling was reduced. Interestingly, E2 and estrogenic compounds showed distinct effects on the contractile activity of uterine cells. E2 enhanced the contractility, while OP and BPA significantly decreased it. Immunohistochemical analysis of CAPs showed distinct regulation of prostaglandin F receptor localization by E2 and estrogenic compounds, which may explain the different contractile activities of those reagents. In summary, we demonstrate that E2, OP, and BPA regulate CAP expression in a similar manner in the immature rat uterus, however, the effects on contractile activity were modulated differently. These findings suggest that OP and BPA interfere with uterine contractility.
Collapse
Affiliation(s)
- Beum-Soo An
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea.
| | | | | | | | | | | | | |
Collapse
|
21
|
Zhang Y, Knutsen GR, Brown MD, Ruest LB. Control of endothelin-a receptor expression by progesterone is enhanced by synergy with Gata2. Mol Endocrinol 2013; 27:892-908. [PMID: 23592430 PMCID: PMC3656236 DOI: 10.1210/me.2012-1334] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 04/10/2013] [Indexed: 11/19/2022] Open
Abstract
The endothelin-A receptor (Ednra) is involved in several physiological, pathological, and developmental pathways. Known for its function in vasoconstriction after being activated by endothelin-1, Ednra also controls cephalic neural crest cell development and appears to play a role in several pathologies, including cancer and periodontitis. However, the mechanisms regulating Ednra expression have not been identified despite its important functions. In this study, we investigated the role progesterone plays in Ednra gene expression in vivo and in vitro. In mice, pregnancy promotes Ednra expression in the heart, kidney, lung, uterus, and placenta, and the up-regulation is mediated by progesterone. We determined that the conserved region between -5.7 and -4.2 kb upstream of the mouse Ednra gene is necessary for the progesterone response. We also found that progesterone mediates Ednra activation through progesterone receptor B activation by its recruitment to PRE6, one of the 6 progesterone response elements found in that locus. However, gene activation by means of a GATA2 site was also necessary for the progesterone response. The Gata2 transcription factor enhances the progesterone response mediated by the progesterone receptor B. Together these results indicate that progesterone regulates Ednra expression by synergizing with Gata2 activity, a previously unknown mechanism. This mechanism may have an impact on pathologies involving the endothelin signaling.
Collapse
Affiliation(s)
- Yanping Zhang
- Center for Craniofacial Research and Diagnosis and Department of Biomedical Sciences, Texas A&M University-Baylor College of Dentistry, Dallas, Texas 75246, USA
| | | | | | | |
Collapse
|
22
|
miR-200 family and targets, ZEB1 and ZEB2, modulate uterine quiescence and contractility during pregnancy and labor. Proc Natl Acad Sci U S A 2010; 107:20828-33. [PMID: 21079000 DOI: 10.1073/pnas.1008301107] [Citation(s) in RCA: 219] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Throughout most of pregnancy, uterine quiescence is maintained by increased progesterone receptor (PR) transcriptional activity, whereas spontaneous labor is initiated/facilitated by a concerted series of biochemical events that activate inflammatory pathways and have a negative impact on PR function. In this study, we uncovered a previously undescribed regulatory pathway whereby micro-RNAs (miRNAs) serve as hormonally modulated and conserved mediators of contraction-associated genes in the pregnant uterus in the mouse and human. Using miRNA and gene expression microarray analyses of uterine tissues, we identified a conserved family of miRNAs, the miR-200 family, that is highly induced at term in both mice and humans as well as two coordinately down-regulated targets, zinc finger E-box binding homeobox proteins ZEB1 and ZEB2, which act as transcriptional repressors. We also observed up-regulation of the miR-200 family and down-regulation of ZEB1 and ZEB2 in two different mouse models of preterm labor. We further demonstrated that ZEB1 is directly up-regulated by the action of progesterone (P(4))/PR at the ZEB1 promoter. Excitingly, we observed that ZEB1 and ZEB2 inhibit expression of the contraction-associated genes, oxytocin receptor and connexin-43, and block oxytocin-induced contractility in human myometrial cells. Together, these findings implicate the miR-200 family and their targets, ZEB1 and ZEB2, as unique P(4)/PR-mediated regulators of uterine quiescence and contractility during pregnancy and labor and shed light on the molecular mechanisms involved in preterm birth.
Collapse
|
23
|
Romero R, Friel LA, Velez Edwards DR, Kusanovic JP, Hassan SS, Mazaki-Tovi S, Vaisbuch E, Kim CJ, Erez O, Chaiworapongsa T, Pearce BD, Bartlett J, Salisbury BA, Anant MK, Vovis GF, Lee MS, Gomez R, Behnke E, Oyarzun E, Tromp G, Williams SM, Menon R. A genetic association study of maternal and fetal candidate genes that predispose to preterm prelabor rupture of membranes (PROM). Am J Obstet Gynecol 2010; 203:361.e1-361.e30. [PMID: 20673868 DOI: 10.1016/j.ajog.2010.05.026] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Revised: 04/10/2010] [Accepted: 05/18/2010] [Indexed: 01/19/2023]
Abstract
OBJECTIVE We sought to determine whether maternal/fetal single-nucleotide polymorphisms (SNPs) in candidate genes are associated with preterm prelabor rupture of membranes (pPROM). STUDY DESIGN A case-control study was conducted in patients with pPROM (225 mothers and 155 fetuses) and 599 mothers and 628 fetuses with a normal pregnancy; 190 candidate genes and 775 SNPs were studied. Single locus/haplotype association analyses were performed; false discovery rate was used to correct for multiple testing (q* = 0.15). RESULTS First, a SNP in tissue inhibitor of metalloproteinase 2 in mothers was significantly associated with pPROM (odds ratio, 2.12; 95% confidence interval, 1.47-3.07; P = .000068), and this association remained significant after correction for multiple comparisons. Second, haplotypes for Alpha 3 type IV collagen isoform precursor in the mother were associated with pPROM (global P = .003). Third, multilocus analysis identified a 3-locus model, which included maternal SNPs in collagen type I alpha 2, defensin alpha 5 gene, and endothelin 1. CONCLUSION DNA variants in a maternal gene involved in extracellular matrix metabolism doubled the risk of pPROM.
Collapse
Affiliation(s)
- Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development/National Institutes of Health/Department of Health and Human Services, Bethesda, MD, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Breuiller-Fouché M, Dubois O, Sediki M, Garcia-Verdugo I, Palaniyar N, Tanfin Z, Chissey A, Cabrol D, Charpigny G, Mehats C. Secreted surfactant protein A from fetal membranes induces stress fibers in cultured human myometrial cells. Am J Physiol Endocrinol Metab 2010; 298:E1188-97. [PMID: 20233942 DOI: 10.1152/ajpendo.00746.2009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the present study, we investigated the ability of human fetal membranes (amnion and choriodecidua) to regulate human maternal uterine cell functions through the secretion of surfactant protein (SP)-A and SP-D at the end of pregnancy. We detected the expression of both SP-A (SP-A1 and SP-A2) and SP-D by quantitative reverse transcription polymerase chain reaction. Immunohistochemistry revealed that human fetal membranes expressed both SP-A and SP-D. By Western blot analysis, we demonstrated that SP-A protein expression was predominant in choriodecidua, whereas the amnion predominantly expressed SP-D. Only the secretion of SP-A was evidenced in the culture supernatants of amnion and choriodecidua explants by immunodot blot and confirmed by Western blot. Exogenous human purified SP-A induced stress fiber formation in cultured human myometrial cells via a pathway involving Rho-kinase. Conditioned medium from choriodecidua and amnion explants mimicked the SP-A effect. Treatment of myometrial cells with SP-A-depleted conditioned medium from choriodecidua or amnion explants failed to change the actin dynamic. These data indicate that SP-A released by human fetal membranes is able to exert a paracrine regulation of F-actin filament organization in myometrial cells.
Collapse
Affiliation(s)
- Michelle Breuiller-Fouché
- Institut National de la Santé et de la Recherche Médicale (INSERM), U767, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Paris V, Paris, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Fitzgibbon J, Morrison JJ, Smith TJ, O'Brien M. Modulation of human uterine smooth muscle cell collagen contractility by thrombin, Y-27632, TNF alpha and indomethacin. Reprod Biol Endocrinol 2009; 7:2. [PMID: 19133144 PMCID: PMC2645409 DOI: 10.1186/1477-7827-7-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2008] [Accepted: 01/08/2009] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Preterm labour occurs in approximately 10% of pregnancies and is a major cause of infant morbidity and mortality. However, the pathways involved in regulating contractility in normal and preterm labour are not fully elucidated. Our aim was to utilise a human myometrial contractility model to investigate the effect of a number of uterine specific contractility agents in this system. Therefore, we investigated the contractile response of human primary uterine smooth muscle cells or immortalised myometrial smooth muscle cells cultured within collagen lattices, to known mediators of uterine contractility, which included thrombin, the ROCK-1 inhibitor Y-27632, tumour necrosis factor alpha (TNF alpha) and the non-steroidal anti-inflammatory indomethacin. METHODS Cell contractility was calculated over time, with the collagen gel contraction assay, utilising human primary uterine smooth muscle cells (hUtSMCs) and immortalised myometrial smooth muscle cells (hTERT-HM): a decrease in collagen gel area equated to an increase in contractility. RNA was isolated from collagen embedded cells and gene expression changes were analysed by real time fluorescence reverse transcription polymerase chain reaction. Scanning electron and fluorescence microscopy were employed to observe cell morphology and cell collagen gel interactions. Statistical analysis was performed using ANOVA followed by Tukey's post hoc tests. RESULTS TNF alpha increased collagen contractility in comparison to the un-stimulated collagen embedded hUtSMC cells, which was inhibited by indomethacin, while indomethacin alone significantly inhibited contraction. Thrombin augmented the contractility of uterine smooth muscle cell and hTERT-HM collagen gels, this effect was inhibited by the thrombin specific inhibitor, hirudin. Y-27632 decreased both basal and thrombin-induced collagen contractility in the hTERT-HM embedded gels. mRNA expression of the thrombin receptor, F2R was up-regulated in hUtSMCs isolated from collagen gel lattices, following thrombin-stimulated contractility. CONCLUSION TNF alpha and thrombin increased uterine smooth muscle cell collagen contractility while indomethacin had the opposite effect. Thrombin-induced collagen contractility resulted in F2R activation which may in part be mediated by the ROCK-1 pathway. This study established the in vitro human myometrial model as a viable method to assess the effects of a range of uterotonic or uterorelaxant agents on contractility, and also permits investigation of the complex regulatory pathways involved in mediating myometrial contractility at labour.
Collapse
Affiliation(s)
- Joan Fitzgibbon
- National Centre for Biomedical and Engineering Science, Orbsen Building, National University of Ireland Galway, University Road, Galway, Ireland
| | - John J Morrison
- National Centre for Biomedical and Engineering Science, Orbsen Building, National University of Ireland Galway, University Road, Galway, Ireland
- Department of Obstetrics and Gynaecology, Clinical Science Institute, University College Hospital Galway, Newcastle Road, Galway, Ireland
| | - Terry J Smith
- National Centre for Biomedical and Engineering Science, Orbsen Building, National University of Ireland Galway, University Road, Galway, Ireland
| | - Margaret O'Brien
- National Centre for Biomedical and Engineering Science, Orbsen Building, National University of Ireland Galway, University Road, Galway, Ireland
| |
Collapse
|
26
|
Tamama K, Sen CK, Wells A. Differentiation of bone marrow mesenchymal stem cells into the smooth muscle lineage by blocking ERK/MAPK signaling pathway. Stem Cells Dev 2008; 17:897-908. [PMID: 18564029 DOI: 10.1089/scd.2007.0155] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Smooth muscle cells (SMCs) are major components of blood vessels and other hollow visceral organs required for tissue engineering of these organs. This study aims to evaluate whether adult bone marrow-derived mesenchymal stem cells (BMMSCs), multipotent cells, can be converted into SMCs. We examined the ERK/MAPK pathway as it exerts anti-myogenic signals in SMCs. Undifferentiated BMMSCs express most SMC marker genes, albeit mainly at low levels, except smooth muscle myosin heavy chain (SMMHC), the most definitive marker of differentiated SMC. The treatment of BMMSC with MEK inhibitor up-regulated the expression of alpha-smooth muscle actin (ASMA), h-caldesmon, and SMMHC in BMMSC in low serum condition. MEK inhibitor-treated BMMSC also contracted a collagen gel in response to endothelin. Interestingly, inhibition of MEK induced myocardin expression in BMMSC. In conclusion, BMMSCs treated MEK inhibitor gain a SMC-like phenotype with ligand-induced cell contractility to endothelin in vitro. This approach has obvious implications for cell therapeutics and tissue engineering of hollow visceral organs such as blood vessels.
Collapse
Affiliation(s)
- Kenichi Tamama
- Department of Pathology, Ohio State University Medical Center, Columbus, Ohio 43210, USA.
| | | | | |
Collapse
|
27
|
O'Brien M, Morrison JJ, Smith TJ. Upregulation of PSCDBP, TLR2, TWIST1, FLJ35382, EDNRB, and RGS12 gene expression in human myometrium at labor. Reprod Sci 2008; 15:382-93. [PMID: 18497345 DOI: 10.1177/1933719108316179] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The regulatory mechanisms underlying myometrial smooth muscle contractility during labor are poorly understood. The authors therefore investigated the transcriptional profile of the changes that occur in the human myometrium at term pregnancy when compared with that at labor. Microarray technology was used to identify differentially expressed genes in human myometrium at labor. Real-time fluorescence reversetranscriptase polymerase chain reaction (RT-PCR) was subsequently performed to verify the microarray data. Semiquantitative RT-PCR, Western blotting, and microscopy methodologies were also used. Certain novel genes were found to be upregulated in human myometrium at labor. Of these, PSCDBP, TLR2, TWIST1 , FLJ35382, andRGS12 have not been previously characterized or identified in human myometrium. EDNRB is the other novel labor-associated gene whose reported expression is also upregulated at labor. All 6 genes were expressed on human myometrial smooth muscle cells. These novel upregulated genes are involved in multiple pathways that may be associated with a variety of cellular processes including inflammation, transcriptional regulation, and intracellular signaling.
Collapse
Affiliation(s)
- Margaret O'Brien
- National Centre for Biomedical and Engineering Science, National University of Ireland Galway, Galway, Ireland.
| | | | | |
Collapse
|
28
|
Garcia-Verdugo I, Tanfin Z, Dallot E, Leroy MJ, Breuiller-Fouché M. Surfactant protein A signaling pathways in human uterine smooth muscle cells. Biol Reprod 2008; 79:348-55. [PMID: 18463356 DOI: 10.1095/biolreprod.108.068338] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The present study investigated the ability of surfactant associated protein A1 (SFTPA1), a major component of lung surfactant, to bind and serve as a signal in human cultured myometrial cells. By using ligand blot analysis with 125I-SFTPA1, we consistently identified two myometrial SFTPA1 interacting proteins (55 and 200 kDa). We found that the SFTPA1 immunoreactive protein was present in myometrial cells. We also showed by indirect immunofluorescence the nuclear translocation of RELA (also known as NFkappaB p65 subunit) after activation of myometrial cells by SFTPA1. Neutralization of TLR4 did not reverse this effect. Moreover, SFTPA1 rapidly activated mitogen-activated protein kinase 1/3 (MAPK1/3) and protein kinase C zeta (PRKCZ). The prolonged treatment of myometrial cells with SFTPA1 upregulated PTGS2 (COX2) protein levels. We next evaluated whether SFTPA1 affected the actin dynamic. Stimulation of myometrial cells with SFTPA1 markedly enhanced the intensity of the filamentous-actin pool stained with fluorescein isothiocyanate-phalloidin. Inhibition of PRKC or Rho-associated, coiled-coil containing protein kinase 1 (ROCK) reduced the SFTPA1-mediated stress fiber formation. Our data support the hypothesis that human myometrial cells express functional SFTPA1 binding sites and respond to SFTPA1 to initiate activation of signaling events related to human parturition.
Collapse
Affiliation(s)
- Ignacio Garcia-Verdugo
- Equipe Signalisation et Régulations Cellulaires, Institut de Biochimie et Biophysique Moléculaire et Cellulaire, CNRS, UMR-8619, Université Paris-Sud, 91400 Orsay, France
| | | | | | | | | |
Collapse
|
29
|
Eude-Le Parco I, Dallot E, Breuiller-Fouché M. Protein kinase C and human uterine contractility. BMC Pregnancy Childbirth 2007; 7 Suppl 1:S11. [PMID: 17570155 PMCID: PMC1892052 DOI: 10.1186/1471-2393-7-s1-s11] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abnormalities in uterine contractility are thought to contribute to several clinical problems, including preterm labor. A better understanding of the mechanisms controlling uterine activity would make it possible to propose more appropriate and effective management practices than those currently in use. Recent advances point to a role of the protein kinase C (PRKC) family in the regulation of uterine smooth muscle contraction at the end of pregnancy. In this review, we highlight recent work that explores the involvement of individual PRKC isoforms in cellular process, with an emphasis on the properties of PRKCZ isoform.
Collapse
Affiliation(s)
| | - Emmanuelle Dallot
- INSERM U767, Paris, F-75006, France
- Université René Descartes, Paris, F-75006, France
| | | |
Collapse
|
30
|
Abstract
One of the classical biological actions mediated by the posterior pituitary hormone oxytocin (OT) is contraction of the uterus at parturition. Moreover, premature activation of the OT system is thought to contribute to preterm labor, a major clinical problem in obstetrical practice. However, the molecular mechanisms linking activation of the OT receptor (OTR) to myometrial contractions are not fully understood. Here, we describe an in vitro system that should serve as a useful tool to study this question at a cellular level. The system consists of a collagen lattice contraction assay and two different human myometrial cell lines: a cell clone from a telomerase-immortalized human myometrial cell population (hTERT-C3) as well as a cell line derived from a primary culture of human myometrial cells (M11). Using this approach, we observed that 1 nM OT promoted an almost maximal effect on cell contraction in both cell lines tested. Furthermore, this dose-dependent, OT-induced contraction was antagonized by the specific OTR antagonist d(CH(2))(5)[Tyr(Me)(2),Thr(4),Tyr-NH(2)(9)]OVT as well as the clinically used antagonist atosiban. This cell line-based contraction assay enables the application of molecular tools aimed at suppressing or overexpressing specific genes. It is also amenable to high-throughput testing approaches. Therefore, this system represents a powerful and improved experimental model that should facilitate the study of the molecular signal transduction pathways involved in the uterotonic actions of OT.
Collapse
Affiliation(s)
- Dominic Devost
- Dept. of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montreal, QC, H3G 1Y6, Canada
| | | |
Collapse
|
31
|
Ivanov V, Roomi MW, Kalinovsky T, Niedzwiecki A, Rath M. Natural nutrient mixture effectively reduces collagen matrix contraction driven by human uterine smooth muscle cells. J Obstet Gynaecol Res 2006; 32:23-31. [PMID: 16445522 DOI: 10.1111/j.1447-0756.2006.00367.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIM Abnormal uterine myometrial contractility causes preterm delivery, contributing to perinatal morbidity and mortality. Disturbances in hormonal regulation and inflammation-related processes have been attributed a role in the pathophysiological mechanisms of uterine contractility. We investigated the effects of natural nutrients on uterine tissue contractility in vitro. METHODS We used an in vitro model of collagen I gel contraction driven by embedded cultured human uterine smooth muscle cells (SMC). The effects of tested compounds were evaluated using their capacity to affect gel contraction (measured by reduction in gel area during 24-h incubation in serum free medium). Cellular expression of matrix metalloproteinases (MMP) was followed by gel zymography. RESULTS Collagen gel contraction driven by uterine SMC was significantly stimulated by potassium chloride, pituitary hormone oxytocin and by inflammatory cytokine alpha-tumor necrosis factor (TNF-alpha). Accelerated gel contraction was accompanied by elevated secretion of MMP-2 and MMP-9 into cell culture media. Among a variety of purified bioflavonoids and polyphenol-enriched plant extracts tested for their ability to counteract uterine SMC-dependent collagen gel contraction, the strongest effects were demonstrated by epigallocatechin gallate and green tea leaf extract, respectively. The addition of ascorbic acid and the amino acids lysine, arginine, cysteine and proline to green tea extract further increased its effectiveness. A reduction in gel contraction correlated with decreased MMP expression. CONCLUSION Based on these findings, we found that nutrients can effectively counteract uterine myometrial contraction and MMP activity in vitro, suggesting that pathophysiological mechanisms of abnormal uterine myometrial contractility can be counteracted by a combination of naturally occurring nutrients. These mechanisms might involve extracellular matrix remodeling.
Collapse
Affiliation(s)
- Vadim Ivanov
- Matthias Rath Research Institute, Santa Clara, California, USA
| | | | | | | | | |
Collapse
|
32
|
Breuiller-Fouché M, Morinière C, Dallot E, Oger S, Rebourcet R, Cabrol D, Leroy MJ. Regulation of the endothelin/endothelin receptor system by interleukin-1{beta} in human myometrial cells. Endocrinology 2005; 146:4878-86. [PMID: 16109787 DOI: 10.1210/en.2005-0250] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Proinflammatory cytokines produced at the fetomaternal interface, such as IL-1beta, have been implicated in preterm and term labor. The present study was performed to evaluate the influence of IL-1beta on the endothelin (ET)/ET receptor system in human myometrial cells. We report that myometrial cells under basal conditions not only respond to but also secrete ET-1, one of the main regulators of uterine contractions. Prolonged exposure of the cells to IL-1beta led to a decrease in prepro-ET-1 and ET-3 mRNA correlated with a decrease in immunoreactive ET-1 and ET-3 levels in the culture medium. Whereas ETA receptor expression at both protein and mRNA levels was not affected by IL-1beta treatment, we demonstrated an unexpected predominance of the ETB receptor subtype under this inflammatory condition. Whereas the physiological function of ETB remains unclear, we confirmed that only ETA receptors mediate ET-1-induced myometrial cell contractions under basal conditions. By contrast, prolonged exposure of the cells to IL-1beta abolished the contractile effect induced by ET-1. Such a regulation of IL-1beta on the ET release and the balance of ETA to ETB receptors leading to a loss of ET-1-induced myometrial cell contractions suggest that complex regulatory mechanisms take place to constraint the onset of infection-induced premature contractions.
Collapse
|
33
|
Cellini M, Versura P, Trerè D, Campos EC. Effects of Endothelin-1 on Human Trabecular Meshwork Cell Contraction. Ophthalmic Res 2005; 37:43-9. [PMID: 15637421 DOI: 10.1159/000083021] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2003] [Accepted: 08/11/2004] [Indexed: 12/31/2022]
Abstract
Trabecular meshwork (TM) cells are now considered to play an active role in the aqueous outflow mechanism, since they exhibit smooth muscle-like contractile properties. Endothelin-1 (ET-1), a potent vasoconstrictor peptide, has been proposed to play a role in the local regulation of aqueous outflow and intra-ocular pressure control. We propose an in vitro culture model as a method in the study of ET-1-induced human trabecular meshwork (HTM) cell contractility. Experiments were performed on semi-confluent HTM cells (primary cultures established from normotensive human donor eyes) at the 2nd passage with PBS as control, ET-1, sarafotoxin 6c (a selective endothelin B receptor agonist) and Y-27632 (a selective inhibitor of rho-associated kinase). The contractile status of the cells was evaluated by a morphometric analysis of the cell area, assuming that HTM cells in culture are able to reduce their area as a consequence of cytoskeletal contraction rather than regulatory volume decrease. Our data indicate that image analysis of the HTM cell area can be a reliable method in the study of TM cell contractility, since it utilizes human cells and offers versatile opportunities for the pharmacological evaluation of drugs controlling HTM cell status.
Collapse
Affiliation(s)
- Mauro Cellini
- Department of Surgery and Transplant, Section of Ophthalmology I, Alma Mater Studiorum University of Bologna, Bologna, Italy.
| | | | | | | |
Collapse
|
34
|
Méhats C, Oger S, Leroy MJ. Cyclic nucleotide phosphodiesterase-4 inhibitors: a promising therapeutic approach to premature birth? Eur J Obstet Gynecol Reprod Biol 2005; 117 Suppl 1:S15-7. [PMID: 15530708 DOI: 10.1016/j.ejogrb.2004.07.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Pharmacological interventions in preterm labour attempt to target various protagonists involved in the signalling pathway leading to myometrial contractions. None of these interventions has clearly demonstrated any real clinical benefit, and some are associated with severe maternal and fetal side effects. The original study now reported has disclosed a new potential target, myometrial phosphodiesterase-4 (PDE4), for tocolysis. The PDE4 family specifically hydrolyses cAMP, a major intracellular second messenger implicated in the mechanisms governing smooth muscle motility. One particular isoform, PDE4B2, is overexpressed in the human myometrium at the end of pregnancy, and we observed a change of the myorelaxant properties of selective PDE4 inhibitors at this time. Signalling factors involved in labour at term or in infection-induced preterm labour, such as PGE2 or IL-1beta, are also able to induce PDE4B2 expression by way of a cAMP-dependent pathway. This feedback loop explains, at least in part, the process of desensitisation to relaxant agents that occurs at the end of pregnancy. In addition, PDE4 inhibitors block the production of LPS-induced inflammatory cytokines by myometrial explants. Thus, a combination of myorelaxant and anti-inflammatory properties make the PDE4 inhibitors particularly attractive as a target for the prevention of threatened infection-induced preterm delivery and its consequences for the premature infant.
Collapse
Affiliation(s)
- Céline Méhats
- INSERM U361, Pavillon Baudelocque, Maternité Port-Royal Cochin, 123 Blvd Port Royal, 75014 Paris, France
| | | | | |
Collapse
|
35
|
Di Liberto G, Dallot E, Eude-Le Parco I, Cabrol D, Ferré F, Breuiller-Fouché M. A critical role for PKC zeta in endothelin-1-induced uterine contractions at the end of pregnancy. Am J Physiol Cell Physiol 2003; 285:C599-607. [PMID: 12748064 DOI: 10.1152/ajpcell.00040.2003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have previously shown that protein kinase C (PKC) zeta and/or PKC delta are necessary for endothelin-1 (ET-1)-induced human myometrial contraction at the end of pregnancy (Eude I, Paris P, Cabrol D, Ferré F, and Breuiller-Fouché M. Biol Reprod 63: 1567-1573, 2000). Here, we report that the selective inhibitor of PKC delta isoform, Rottlerin, does not prevent ET-1-induced contractions, whereas LY-294002, a phosphatidylinositol (PI) 3-kinase inhibitor, affects the contractile response. This study characterized the in vitro contractile response of cultured human pregnant myometrial cells to ET-1 known to induce in vitro contractions of intact uterine smooth muscle strips. Cultured myometrial cells incorporated into collagen lattices have the capacity to reduce the size of these lattices, referred to as lattice contraction. Neither the selective conventional PKC isoform inhibitor, Gö-6976, or rottlerin affected myometrial cell-mediated gel contraction by ET-1, whereas this effect was blocked by LY-294002. We found that treatment of myometrial cell lattices with an inhibitory peptide specific for PKC zeta or with an antisense against PKC zeta resulted in a significant loss of ET-1-induced contraction. Evidence is also presented by using confocal microscopy that ET-1 induced translocation of PKC zeta to a structure coincident with the actin-rich microfilaments of the cytoskeleton. We have shown that PKC zeta has a role in the actin organization in ET-1-stimulated cells. Accordingly, our results suggest that PKC zeta plays a role in myometrial contraction in pregnant women.
Collapse
Affiliation(s)
- G Di Liberto
- Institut National de la Santé et de la Recherche Médicale U361, Pavillon Baudelocque, 75014 Paris, France
| | | | | | | | | | | |
Collapse
|