1
|
Bigger-Allen A, Gheinani AH, Adam RM. Investigation of the impact of bromodomain inhibition on cytoskeleton stability and contraction. Cell Commun Signal 2024; 22:184. [PMID: 38493137 PMCID: PMC10944605 DOI: 10.1186/s12964-024-01553-6] [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: 12/21/2023] [Accepted: 03/01/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Injury to contractile organs such as the heart, vasculature, urinary bladder and gut can stimulate a pathological response that results in loss of normal contractility. PDGF and TGFβ are among the most well studied initiators of the injury response and have been shown to induce aberrant contraction in mechanically active cells of hollow organs including smooth muscle cells (SMC) and fibroblasts. However, the mechanisms driving contractile alterations downstream of PDGF and TGFβ in SMC and fibroblasts are incompletely understood, limiting therapeutic interventions. METHODS To identify potential molecular targets, we have leveraged the analysis of publicly available data, comparing transcriptomic changes in mechanically active cells stimulated with PDGF and TGFβ. Additional Analysis of publicly available data sets were performed on SMC and fibroblasts treated in the presence or absence of the MYC inhibitor JQ1. Validation of in silico findings were performed with qPCR, immunoblots, and collagen gel contraction assays measure the effect of JQ1 on cytoskeleton associated genes, proteins and contractility in mechanically active cells. Likelihood ratio test and FDR adjusted p-values were used to determine significant differentially expressed genes. Student ttest were used to calculate statistical significance of qPCR and contractility analyses. RESULTS Comparing PDGF and TGFβ stimulated SMC and fibroblasts identified a shared molecular profile regulated by MYC and members of the AP-1 transcription factor complex. Additional in silico analysis revealed a unique set of cytoskeleton-associated genes that were sensitive to MYC inhibition with JQ1. In vitro validation demonstrated JQ1 was also able to attenuate TGFβ and PDGF induced changes to the cytoskeleton and contraction of smooth muscle cells and fibroblasts in vitro. CONCLUSIONS These findings identify MYC as a key driver of aberrant cytoskeletal and contractile changes in fibroblasts and SMC, and suggest that JQ1 could be used to restore normal contractile function in hollow organs.
Collapse
Affiliation(s)
- Alexander Bigger-Allen
- Urological Diseases Research Center, Boston Children's Hospital, Enders Bldg 1061.4, 300 Longwood Avenue, Boston, MA, 02115, USA
- Biological & Biomedical Sciences Program, Division of Medical Sciences, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ali Hashemi Gheinani
- Urological Diseases Research Center, Boston Children's Hospital, Enders Bldg 1061.4, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Functional Urology Research Group, Department for BioMedical Research DBMR, University of Bern, Bern, Switzerland
- Department of Urology, Inselspital University Hospital, 3010, Bern, Switzerland
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rosalyn M Adam
- Urological Diseases Research Center, Boston Children's Hospital, Enders Bldg 1061.4, 300 Longwood Avenue, Boston, MA, 02115, USA.
- Department of Surgery, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
2
|
Bigger-Allen A, Gheinani AH, Adam RM. Investigation of the impact of bromodomain inhibition on cytoskeleton stability and contraction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.14.567076. [PMID: 38014184 PMCID: PMC10680757 DOI: 10.1101/2023.11.14.567076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Injury to contractile organs such as the heart, vasculature, urinary bladder and gut can stimulate a pathological response that results in loss of normal contractility. PDGF and TGFβ are among the most well studied initiators of the injury response and have been shown to induce aberrant contraction in mechanically active cells of hollow organs including smooth muscle cells (SMC) and fibroblasts. However the mechanisms driving contractile alterations downstream of PDGF and TGFβ in SMC and fibroblasts are incompletely understood, limiting therapeutic interventions. To identify potential molecular targets, we have leveraged the analysis of publicly available data, comparing transcriptomic changes in mechanically active cells stimulated with PDGF and TGFβ and identified a shared molecular profile regulated by MYC and members of the AP-1 transcription factor complex. We also analyzed data sets from SMC and fibroblasts treated in the presence or absence of the MYC inhibitor JQ1. This analysis revealed a unique set of cytoskeleton-associated genes that were sensitive to MYC inhibition. JQ1 was also able to attenuate TGFβ and PDGF induced changes to the cytoskeleton and contraction of smooth muscle cells and fibroblasts in vitro. These findings identify MYC as a key driver of aberrant cytoskeletal and contractile changes in fibroblasts and SMC, and suggest that JQ1 could be used to restore normal contractile function in hollow organs.
Collapse
Affiliation(s)
- Alexander Bigger-Allen
- Urological Diseases Research Center, Boston Children’s Hospital, Boston, MA, USA
- Biological & Biomedical Sciences Program, Division of Medical Sciences, Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ali Hashemi Gheinani
- Urological Diseases Research Center, Boston Children’s Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Functional Urology Research Group, Department for BioMedical Research DBMR, University of Bern, Switzerland
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rosalyn M. Adam
- Urological Diseases Research Center, Boston Children’s Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
3
|
Kim JO, Baek SE, Jeon EY, Choi JM, Jang EJ, Kim CD. PDGFR-β signaling mediates HMGB1 release in mechanically stressed vascular smooth muscle cells. PLoS One 2022; 17:e0265191. [PMID: 35294955 PMCID: PMC8926240 DOI: 10.1371/journal.pone.0265191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 02/18/2022] [Indexed: 11/19/2022] Open
Abstract
Mechanically stressed vascular smooth muscle cells (VSMCs) have potential roles in the development of vascular complications. However, the underlying mechanisms are unclear. Using VSMCs cultured from rat thoracic aorta explants, we investigated the effects of mechanical stretch (MS) on the cellular secretion of high mobility group box 1 (HMGB1), a major damage-associated molecular pattern that mediates vascular complications in stressed vasculature. Enzyme-linked immunosorbent assay (ELISA) demonstrated an increase in the secretion of HMGB1 in VSMCs stimulated with MS (0–3% strain, 60 cycles/min), and this secretion was markedly and time-dependently increased at 3% MS. The increased secretion of HMGB1 at 3% MS was accompanied by an increased cytosolic translocation of nuclear HMGB1; the acetylated and phosphorylated forms of this protein were significantly increased. Among various inhibitors of membrane receptors mediating mechanical signals, AG1295 (a platelet-derived growth factor receptor (PDGFR) inhibitor) attenuated MS-induced HMGB1 secretion. Inhibitors of other receptors, including epidermal growth factor, insulin-like growth factor, and fibroblast growth factor receptors, did not inhibit this secretion. Additionally, MS-induced HMGB1 secretion was markedly attenuated in PDGFR-β-deficient cells but not in cells transfected with PDGFR-α siRNA. Likewise, PDGF-DD, but not PDGF-AA, directly increased HMGB1 secretion in VSMCs, indicating a pivotal role of PDGFR-β signaling in the secretion of this protein in VSMCs. Thus, targeting PDGFR-β-mediated secretion of HMGB1 in VSMCs might be a promising therapeutic strategy for vascular complications associated with hypertension.
Collapse
Affiliation(s)
- Ji On Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Seung Eun Baek
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Eun Yeong Jeon
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Jong Min Choi
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Eun Jeong Jang
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Chi Dae Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- * E-mail:
| |
Collapse
|
4
|
Xie W, Zhang L, Luo W, Zhai Z, Wang C, Shen YH. AKT2 regulates endothelial-mediated coagulation homeostasis and promotes intrathrombotic recanalization and thrombus resolution in a mouse model of venous thrombosis. J Thromb Thrombolysis 2021; 50:98-111. [PMID: 32358666 DOI: 10.1007/s11239-020-02112-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Venous thromboembolism (VTE) carries a high risk of morbidity and mortality. Understanding the mechanisms of venous thrombus formation and resolution is critical for improving VTE management. AKT2 kinase is essential for platelet activation and arterial thrombosis. In this study, we examined the role of AKT2 in venous thrombosis in a mouse model of venous thrombosis induced by inferior vena cava (IVC) ligation. We observed an induction of AKT2 expression in the ligated IVC of wild-type (WT) mice. Interestingly, although the initial thrombus size of the ligated IVC was similar between Akt2-/- mice and WT mice, thrombus resolution was delayed in the ligated IVC of Akt2-/- mice. Compared with the ligated IVC of WT mice, the ligated IVC of Akt2-/- mice displayed decreased levels of thrombomodulin (TM) and increased levels of tissue factor (TF), apoptosis, and necroptosis. In addition, intrathrombotic endothelial cells in the ligated IVC of Akt2-/- mice failed to form small vessels, resulting in impaired recanalization and thrombus resolution. TGF-β signaling activation and fibrotic remodeling were increased in the thrombus and vein wall of the ligated IVC of Akt2-/- mice. We further investigated the AKT2-mediated regulation of coagulation factors in endothelial cells and found that forkhead box protein O1 (FOXO1), a target of AKT, enhanced TF and inhibited TM expression. By inhibiting FOXO1, AKT2 suppressed TF expression while increasing TM expression. Our findings indicate that AKT2 may protect endothelial cells against cell death, regulate endothelial-mediated coagulation homeostasis, and promote intrathrombotic recanalization and thrombus resolution in venous thrombosis. These observations suggest dynamic roles of AKT2 in venous thrombus formation and resolution.
Collapse
Affiliation(s)
- Wanmu Xie
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, BCM 390, Houston, TX, 77030, USA.,National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Lin Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, BCM 390, Houston, TX, 77030, USA.,Texas Heart Institute, Houston, TX, USA
| | - Wei Luo
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, BCM 390, Houston, TX, 77030, USA.,Texas Heart Institute, Houston, TX, USA
| | - Zhenguo Zhai
- Department of Respiratory and Critical Care Medicine, China-Japan Friendship Hospital, 2 Yinghua Dongjie, Hepingli, Beijing, 100029, China.,Department of Respiratory Medicine, Capital Medical University, Beijing, China
| | - Chen Wang
- National Clinical Research Center for Respiratory Diseases, Beijing, China. .,Department of Respiratory and Critical Care Medicine, China-Japan Friendship Hospital, 2 Yinghua Dongjie, Hepingli, Beijing, 100029, China. .,Department of Respiratory Medicine, Capital Medical University, Beijing, China.
| | - Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, BCM 390, Houston, TX, 77030, USA. .,Texas Heart Institute, Houston, TX, USA.
| |
Collapse
|
5
|
Wang KC, Chen PS, Chao TH, Luo CY, Chung HC, Tseng SY, Huang TY, Lin YL, Shi GY, Wu HL, Li YH. The role of vascular smooth muscle cell membrane-bound thrombomodulin in neointima formation. Atherosclerosis 2019; 287:54-63. [PMID: 31212235 DOI: 10.1016/j.atherosclerosis.2019.05.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 04/24/2019] [Accepted: 05/23/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND AIMS Thrombomodulin (TM) is an endothelial cell membrane-bound anticoagulant protein expressed in normal arteries. After vascular injury, medial and neointimal smooth muscle cells (SMCs) exhibit large amounts of TM. The purpose of this study was to investigate the physiological significance of vascular SMC-bound TM. METHODS The morphology, expression of phenotype markers and cell behaviors of cultured aortic SMCs after knockdown of TM were observed. Transgenic mice with SMC-specific TM deletion were generated, and carotid neointima formation was induced by carotid ligation. RESULTS Cultured human aortic SMCs displayed a synthetic phenotype with a rhomboid-shaped morphology and expressed TM. TM knockdown induced a spindle-shaped change in morphology with an increased expression of contractile phenotype marker and decreased expression of synthetic phenotype marker. TM knockdown not only attenuated the proliferation of SMCs but also reduced tumor necrosis factor-α-induced nuclear factor-κB activation and interlukin-6 production. In a carotid artery ligation model, transgenic mice with SMC-specific TM deletion (SM22-cretg/TMflox/flox) had significantly less cellular proliferation in arterial walls compared with wild type mice (SM22-cretg/TM+/+). The neointima area and neointima/media area ratio were smaller in SM22-cretg/TMflox/flox mice at 4 weeks after ligation. CONCLUSIONS Our results indicate that vascular SMC-bound TM plays a role in changes of the SMC phenotype. It also influences SMC cell behavior and injury-induced neointima formation.
Collapse
Affiliation(s)
- Kuan-Chieh Wang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Pharmacy, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Po-Sheng Chen
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ting-Hsing Chao
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chawn-Yau Luo
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsing-Chun Chung
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shih-Ya Tseng
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ting-Yu Huang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ying-Li Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Guey-Yueh Shi
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hua-Lin Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Heng Li
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
6
|
Pai VC, Lo IC, Huang YW, Tsai IC, Cheng HP, Shi GY, Wu HL, Jiang MJ. The chondroitin sulfate moiety mediates thrombomodulin-enhanced adhesion and migration of vascular smooth muscle cells. J Biomed Sci 2018; 25:14. [PMID: 29439742 PMCID: PMC5809974 DOI: 10.1186/s12929-018-0415-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 01/30/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Thrombomodulin (TM), a transmembrane glycoprotein highly expressed in endothelial cells (ECs), is a potent anticoagulant maintaining circulation homeostasis. Under inflammatory states, TM expression is drastically reduced in ECs while vascular smooth muscle cells (VSMCs) show a robust expression of TM. The functional role of TM in VSMCs remains elusive. METHODS We examined the role of TM in VSMCs activities in human aortic VSMCs stimulated with platelet-derived growth factor-BB (PDGF-BB). Using rat embryonic aorta-derived A7r5 VSMCs which do not express TM, the role of the chondroitin sulfate (CS) moiety of TM in VSMCs was delineated with cells expressing wild-type TM and the CS-devoid TM mutant. RESULTS Expression of TM enhanced cell migration and adhesion/spreading onto type I collagen, but had no effect on cell proliferation. Knocking down TM with short hairpin RNA reduced PDGF-stimulated adhesion and migration of human aortic VSMCs. In A7r5 cells, TM-mediated cell adhesion was eradicated by pretreatment with chondroitinase ABC which degrades CS moiety. Furthermore, the TM mutant (TMS490, 492A) devoid of CS moiety failed to increase cell adhesion, spreading or migration. Wild-type TM, but not TMS490, 492A, increased focal adhesion kinase (FAK) activation during cell adhesion, and TM-enhanced cell migration was abolished by a function-blocking anti-integrin β1 antibody. CONCLUSION Chondroitin sulfate modification is required for TM-mediated activation of β1-integrin and FAK, thereby enhancing adhesion and migration activity of VSMCs.
Collapse
Affiliation(s)
- Vincent Chunpeng Pai
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, 1 Ta-Hsueh Road, Tainan, 70101, Taiwan
| | - I-Chung Lo
- Cardiovascular Research Center, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Yan Wun Huang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, 1 Ta-Hsueh Road, Tainan, 70101, Taiwan
| | - I-Ching Tsai
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, 1 Ta-Hsueh Road, Tainan, 70101, Taiwan
| | - Hui-Pin Cheng
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, 1 Ta-Hsueh Road, Tainan, 70101, Taiwan
| | - Guey-Yueh Shi
- Cardiovascular Research Center, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.,Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Hua-Lin Wu
- Cardiovascular Research Center, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.,Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Meei Jyh Jiang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, 1 Ta-Hsueh Road, Tainan, 70101, Taiwan. .,Cardiovascular Research Center, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
| |
Collapse
|
7
|
Vasquez E, Cristofaro V, Lukianov S, Burkhard FC, Gheinani AH, Monastyrskaya K, Bielenberg DR, Sullivan MP, Adam RM. Deletion of neuropilin 2 enhances detrusor contractility following bladder outlet obstruction. JCI Insight 2017; 2:e90617. [PMID: 28194441 DOI: 10.1172/jci.insight.90617] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Chronic urethral obstruction and the ensuing bladder wall remodeling can lead to diminished bladder smooth muscle (BSM) contractility and debilitating lower urinary tract symptoms. No effective pharmacotherapy exists to restore BSM contractile function. Neuropilin 2 (Nrp2) is a transmembrane protein that is highly expressed in BSM. Nrp2 deletion in mice leads to increased BSM contraction. We determined whether genetic ablation of Nrp2 could restore BSM contractility following obstruction. Partial bladder outlet obstruction (pBOO) was created by urethral occlusion in mice with either constitutive and ubiquitous, or inducible smooth muscle-specific deletion of Nrp2, and Nrp2-intact littermates. Mice without obstruction served as additional controls. Contractility was measured by isometric tension testing. Nrp2 deletion prior to pBOO increased force generation in BSM 4 weeks following surgery. Deletion of Nrp2 in mice already subjected to pBOO for 4 weeks showed increased contractility of tissues tested 6 weeks after surgery compared with nondeleted controls. Assessment of tissues from patients with urodynamically defined bladder outlet obstruction revealed reduced NRP2 levels in obstructed bladders with compensated compared with decompensated function, relative to asymptomatic controls. We conclude that downregulation of Nrp2 promotes BSM force generation. Neuropilin 2 may represent a novel target to restore contractility following obstruction.
Collapse
Affiliation(s)
- Evalynn Vasquez
- Urological Diseases Research Center, Boston Children's Hospital.,Department of Surgery, Harvard Medical School
| | - Vivian Cristofaro
- Department of Surgery, Harvard Medical School.,Division of Urology, Veterans Affairs Boston Healthcare System, Boston, Massachusetts, USA
| | - Stefan Lukianov
- Urological Diseases Research Center, Boston Children's Hospital
| | - Fiona C Burkhard
- Urology Research Laboratory, Department of Clinical Research, Universität Bern, Bern, Switzerland
| | - Ali Hashemi Gheinani
- Urology Research Laboratory, Department of Clinical Research, Universität Bern, Bern, Switzerland
| | - Katia Monastyrskaya
- Urology Research Laboratory, Department of Clinical Research, Universität Bern, Bern, Switzerland
| | - Diane R Bielenberg
- Department of Surgery, Harvard Medical School.,Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Maryrose P Sullivan
- Department of Surgery, Harvard Medical School.,Division of Urology, Veterans Affairs Boston Healthcare System, Boston, Massachusetts, USA
| | - Rosalyn M Adam
- Urological Diseases Research Center, Boston Children's Hospital.,Department of Surgery, Harvard Medical School
| |
Collapse
|
8
|
Adamowicz J, Pokrywczyńska M, Tworkiewicz J, Kowalczyk T, van Breda SV, Tyloch D, Kloskowski T, Bodnar M, Skopinska-Wisniewska J, Marszałek A, Frontczak-Baniewicz M, Kowalewski TA, Drewa T. New Amniotic Membrane Based Biocomposite for Future Application in Reconstructive Urology. PLoS One 2016; 11:e0146012. [PMID: 26766636 PMCID: PMC4713072 DOI: 10.1371/journal.pone.0146012] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 12/12/2015] [Indexed: 02/07/2023] Open
Abstract
Objective Due to the capacity of the amniotic membrane (Am) to support re-epithelisation and inhibit scar formation, Am has a potential to become a considerable asset for reconstructive urology i.e., reconstruction of ureters and urethrae. The application of Am in reconstructive urology is limited due to a poor mechanical characteristic. Am reinforcement with electrospun nanofibers offers a new strategy to improve Am mechanical resistance, without affecting its unique bioactivity profile. This study evaluated biocomposite material composed of Am and nanofibers as a graft for urinary bladder augmentation in a rat model. Material and Methods Sandwich-structured biocomposite material was constructed from frozen Am and covered on both sides with two-layered membranes prepared from electrospun poly-(L-lactide-co-E-caprolactone) (PLCL). Wistar rats underwent hemicystectomy and bladder augmentation with the biocomposite material. Results Immunohistohemical analysis (hematoxylin and eosin [H&E], anti-smoothelin and Masson’s trichrome staining [TRI]) revealed effective regeneration of the urothelial and smooth muscle layers. Anti-smoothelin staining confirmed the presence of contractile smooth muscle within a new bladder wall. Sandwich-structured biocomposite graft material was designed to regenerate the urinary bladder wall, fulfilling the requirements for normal bladder tension, contraction, elasticity and compliance. Mechanical evaluation of regenerated bladder wall conducted based on Young’s elastic modulus reflected changes in the histological remodeling of the augmented part of the bladder. The structure of the biocomposite material made it possible to deliver an intact Am to the area for regeneration. An unmodified Am surface supported regeneration of the urinary bladder wall and the PLCL membranes did not disturb the regeneration process. Conclusions Am reinforcement with electrospun nanofibers offers a new strategy to improve Am mechanical resistance without affecting its unique bioactivity profile.
Collapse
Affiliation(s)
- Jan Adamowicz
- Chair of Urology, Department of Regenerative Medicine, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, Bydgoszcz, Poland
- Department of General, Oncologic and Pediatric Urology, Nicolaus Copernicus University, Bydgoszcz, Poland
- * E-mail:
| | - Marta Pokrywczyńska
- Chair of Urology, Department of Regenerative Medicine, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, Bydgoszcz, Poland
| | - Jakub Tworkiewicz
- Chair of Urology, Department of Regenerative Medicine, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, Bydgoszcz, Poland
- Department of Urology, Nicolaus Copernicus Hospital Batory, Torun, Poland
| | - Tomasz Kowalczyk
- Laboratory of Modeling in Biology and Medicine, Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
| | - Shane V. van Breda
- Department of Internal Medicine, Division of Infectious Diseases, University of Pretoria, Pretoria, South Africa
| | - Dominik Tyloch
- Chair of Urology, Department of Regenerative Medicine, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, Bydgoszcz, Poland
- Department of General, Oncologic and Pediatric Urology, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Tomasz Kloskowski
- Chair of Urology, Department of Regenerative Medicine, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, Bydgoszcz, Poland
| | - Magda Bodnar
- Department of Clinical Pathomorphology, Faculty of Medicine, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Joanna Skopinska-Wisniewska
- Department of Chemistry of Biomaterials and Cosmetics, Faculty of Chemistry, Nicolaus Copernicus University, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Andrzej Marszałek
- Department of Clinical Pathomorphology, Faculty of Medicine, Nicolaus Copernicus University, Bydgoszcz, Poland
| | | | - Tomasz A. Kowalewski
- Department of Mechanics and Physics of Fluids, Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland, Poland
| | - Tomasz Drewa
- Chair of Urology, Department of Regenerative Medicine, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, Bydgoszcz, Poland
- Department of General, Oncologic and Pediatric Urology, Nicolaus Copernicus University, Bydgoszcz, Poland
- Department of Urology, Nicolaus Copernicus Hospital Batory, Torun, Poland
| |
Collapse
|
9
|
Platelet Derived Growth Factor Has a Role in Pressure Induced Bladder Smooth Muscle Cell Hyperplasia and Acts in a Paracrine Way. J Urol 2015; 194:1797-805. [DOI: 10.1016/j.juro.2015.05.092] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2015] [Indexed: 12/17/2022]
|
10
|
Mauney JR, Adam RM. Dynamic reciprocity in cell-scaffold interactions. Adv Drug Deliv Rev 2015; 82-83:77-85. [PMID: 25453262 DOI: 10.1016/j.addr.2014.10.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 09/07/2014] [Accepted: 10/15/2014] [Indexed: 12/14/2022]
Abstract
Tissue engineering in urology has shown considerable promise. However, there is still much to understand, particularly regarding the interactions between scaffolds and their host environment, how these interactions regulate regeneration and how they may be enhanced for optimal tissue repair. In this review, we discuss the concept of dynamic reciprocity as applied to tissue engineering, i.e. how bi-directional signaling between implanted scaffolds and host tissues such as the bladder drives the process of constructive remodeling to ensure successful graft integration and tissue repair. The impact of scaffold content and configuration, the contribution of endogenous and exogenous bioactive factors, the influence of the host immune response and the functional interaction with mechanical stimulation are all considered. In addition, the temporal relationships of host tissue ingrowth, bioactive factor mobilization, scaffold degradation and immune cell infiltration, as well as the reciprocal signaling between discrete cell types and scaffolds are discussed. Improved understanding of these aspects of tissue repair will identify opportunities for optimization of repair that could be exploited to enhance regenerative medicine strategies for urology in future studies.
Collapse
|
11
|
Integration of proteomic and transcriptomic profiles identifies a novel PDGF-MYC network in human smooth muscle cells. Cell Commun Signal 2014; 12:44. [PMID: 25080971 PMCID: PMC4422302 DOI: 10.1186/s12964-014-0044-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 06/23/2014] [Indexed: 12/12/2022] Open
Abstract
Background Platelet-derived growth factor-BB (PDGF-BB) has been implicated in the proliferation, migration and synthetic activities of smooth muscle cells that characterize physiologic and pathologic tissue remodeling in hollow organs. However, neither the molecular basis of PDGFR-regulated signaling webs, nor the extent to which specific components within these networks could be exploited for therapeutic benefit has been fully elucidated. Results Expression profiling and quantitative proteomics analysis of PDGF-treated primary human bladder smooth muscle cells identified 1,695 genes and 241 proteins as differentially expressed versus non-treated cells. Analysis of gene expression data revealed MYC, JUN, EGR1, MYB, RUNX1, as the transcription factors most significantly networked with up-regulated genes. Forty targets were significantly altered at both the mRNA and protein levels. Proliferation, migration and angiogenesis were the biological processes most significantly associated with this signature, and MYC was the most highly networked master regulator. Alterations in master regulators and gene targets were validated in PDGF-stimulated smooth muscle cells in vitro and in a model of bladder injury in vivo. Pharmacologic inhibition of MYC and JUN confirmed their role in SMC proliferation and migration. Network analysis identified the diaphanous-related formin 3 as a novel PDGF target regulated by MYC and JUN, which was necessary for PDGF-stimulated lamellipodium formation. Conclusions These findings provide the first systems-level analysis of the PDGF-regulated transcriptome and proteome in normal smooth muscle cells. The analyses revealed an extensive cohort of PDGF-dependent biological processes and connected key transcriptional effectors to their regulation, significantly expanding current knowledge of PDGF-stimulated signaling cascades. These observations also implicate MYC as a novel target for pharmacological intervention in fibroproliferative expansion of smooth muscle, and potentially in cancers in which PDGFR-dependent signaling or MYC activation promote tumor progression.
Collapse
|
12
|
Loss of Sh3gl2/endophilin A1 is a common event in urothelial carcinoma that promotes malignant behavior. Neoplasia 2014; 15:749-60. [PMID: 23814487 DOI: 10.1593/neo.121956] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 04/16/2013] [Accepted: 04/22/2013] [Indexed: 11/18/2022] Open
Abstract
Urothelial carcinoma (UC) causes substantial morbidity and mortality worldwide. However, the molecular mechanisms underlying urothelial cancer development and tumor progression are still largely unknown. Using informatics analysis, we identified Sh3gl2 (endophilin A1) as a bladder urothelium-enriched transcript. The gene encoding Sh3gl2 is located on chromosome 9p, a region frequently altered in UC. Sh3gl2 is known to regulate endocytosis of receptor tyrosine kinases implicated in oncogenesis, such as the epidermal growth factor receptor (EGFR) and c-Met. However, its role in UC pathogenesis is unknown. Informatics analysis of expression profiles as well as immunohistochemical staining of tissue microarrays revealed Sh3gl2 expression to be decreased in UC specimens compared to nontumor tissues. Loss of Sh3gl2 was associated with increasing tumor grade and with muscle invasion, which is a reliable predictor of metastatic disease and cancer-derived mortality. Sh3gl2 expression was undetectable in 19 of 20 human UC cell lines but preserved in the low-grade cell line RT4. Stable silencing of Sh3gl2 in RT4 cells by RNA interference 1) enhanced proliferation and colony formation in vitro, 2) inhibited EGF-induced EGFR internalization and increased EGFR activation, 3) stimulated phosphorylation of Src family kinases and STAT3, and 4) promoted growth of RT4 xenografts in subrenal capsule tissue recombination experiments. Conversely, forced re-expression of Sh3gl2 in T24 cells and silenced RT4 clones attenuated oncogenic behaviors, including growth and migration. Together, these findings identify loss of Sh3gl2 as a frequent event in UC development that promotes disease progression.
Collapse
|
13
|
Lin Y, Huang C, Shih C, Chang W, Shyue S, Tsai Y, Lin C, Lee C, Chang Y, Chang N, Lin F, Tsai C. The C-Terminal Domain of Thrombomodulin Regulates Monocyte Migration with Interleukin-6 Stimulation. EUR J INFLAMM 2014. [DOI: 10.1177/1721727x1401200104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Thrombomodulin (TM) is expressed on the surface of monocyte, which is important in the regulation of cell migration, proliferation, and inflammatory responses. In a previous study, we demonstrated that TM on monocyte is negatively associated with cell migration. However, the mechanisms involved in this process are unclear, therefore, we explored the mechanisms in this study. Chemotactic assays and immunofluorescence showed that TM siRNA increased the Chemotaxis of the IL-6-activated THP-1, and aggravated actin assembly relative to the IL-6-treated control. In contrast, cells overexpressing plasmids containing full-length or domain 5 of TM followed by IL-6 treatment displayed lower Chemotaxis and less actin assembly. Western blot analysis showed that TM knockdown markedly increased cytoskeleton components cofilin and LIMK1 phosphorylation in IL-6-treated THP-1, whereas, transfected cells with HA-TM FL or HA-TM D5, but not HA-TM Dl-3 plasmids, reversed the effects. Activation of ERK1/2 and JNK/SAPK, upstream regulators of cytoskeleton components, were also inhibited in overexpressed group. Immunoprecipitation assay demonstrated that actin interacts with TM and intersectin1 in THP-1. Decreased interaction between intersectin1 and actin in TM knockdowns suggested that the interaction is mediated by TM. Our findings indicate that TM domain 5 is a negative regulator and seems to have the ability to inhibit paxillin, cofilin, LIMK1, and actin activation. The mechanisms for the repression effect of domain 5 may be mediated by inhibition of the ERK1/2 and JNK/SAPK activation. Expression of domain 5 of TM may represent a promising approach for controlling monocyte migration, and TM may have potential applications in treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Y.W. Lin
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei
- Division of Cardiovascular Surgery, National Defense Medical Center, Taipei
| | - C.Y. Huang
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei
- Division of Cardiology, Department of Internal Medicine and Taipei Medical University Hospital, Taipei
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei
| | - C.M. Shih
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei
- Division of Cardiology, Department of Internal Medicine and Taipei Medical University Hospital, Taipei
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei
| | - W.L. Chang
- Division of Cardiovascular Surgery, National Defense Medical Center, Taipei
| | - S.K. Shyue
- Institute of Biomedical Sciences, Academia Sinica, Taipei
| | - Y.T. Tsai
- Division of Cardiovascular Surgery, National Defense Medical Center, Taipei
| | - C.Y. Lin
- Division of Cardiovascular Surgery, National Defense Medical Center, Taipei
| | - C.Y. Lee
- Division of Cardiovascular Surgery, National Defense Medical Center, Taipei
| | - Y.J. Chang
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | - N.C. Chang
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei
- Division of Cardiology, Department of Internal Medicine and Taipei Medical University Hospital, Taipei
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei
| | - F.Y. Lin
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei
- Division of Cardiology, Department of Internal Medicine and Taipei Medical University Hospital, Taipei
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei
| | | |
Collapse
|
14
|
Seo KW, Lee SJ, Kim YH, Bae JU, Park SY, Bae SS, Kim CD. Mechanical stretch increases MMP-2 production in vascular smooth muscle cells via activation of PDGFR-β/Akt signaling pathway. PLoS One 2013; 8:e70437. [PMID: 23950935 PMCID: PMC3737227 DOI: 10.1371/journal.pone.0070437] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 06/18/2013] [Indexed: 12/21/2022] Open
Abstract
Increased blood pressure, leading to mechanical stress on vascular smooth muscle cells (VSMC), is a known risk factor for vascular remodeling via increased activity of matrix metalloproteinase (MMP) within the vascular wall. This study aimed to identify cell surface mechanoreceptors and intracellular signaling pathways that influence VSMC to produce MMP in response to mechanical stretch (MS). When VSMC was stimulated with MS (0–10% strain, 60 cycles/min), both production and gelatinolytic activity of MMP-2, but not MMP-9, were increased in a force-dependent manner. MS-enhanced MMP-2 expression and activity were inhibited by molecular inhibition of Akt using Akt siRNA as well as by PI3K/Akt inhibitors, LY293002 and AI, but not by MAPK inhibitors such as PD98059, SP600125 and SB203580. MS also increased Akt phosphorylation in VSMC, which was attenuated by AG1295, a PDGF receptor (PDGFR) inhibitor, but not by inhibitors for other receptor tyrosine kinase including EGF, IGF, and FGF receptors. Although MS activated PDGFR-α as well as PDGFR-β in VSMC, MS-induced Akt phosphorylation was inhibited by molecular deletion of PDGFR-β using siRNA, but not by inhibition of PDGFR-α. Collectively, our data indicate that MS induces MMP-2 production in VSMC via activation of Akt pathway, that is mediated by activation of PDGFR-β signaling pathways.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Gene Expression
- Immunoblotting
- Immunohistochemistry
- Matrix Metalloproteinase 2/genetics
- Matrix Metalloproteinase 2/metabolism
- Microscopy, Confocal
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Phosphorylation
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- RNA Interference
- Rats
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Stress, Mechanical
- Tyrphostins/pharmacology
Collapse
Affiliation(s)
- Kyo Won Seo
- Department of Pharmacology, School of Medicine, and Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Seung Jin Lee
- Department of Pharmacology, School of Medicine, and Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Yun Hak Kim
- Department of Pharmacology, School of Medicine, and Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Jin Ung Bae
- Department of Pharmacology, School of Medicine, and Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - So Youn Park
- Department of Pharmacology, School of Medicine, and Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Sun Sik Bae
- Department of Pharmacology, School of Medicine, and Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Chi Dae Kim
- Department of Pharmacology, School of Medicine, and Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- * E-mail:
| |
Collapse
|
15
|
Ramachandran A, Gangopadhyay SS, Krishnan R, Ranpura SA, Rajendran K, Ram-Mohan S, Mulone M, Gong EM, Adam RM. JunB mediates basal- and TGFβ1-induced smooth muscle cell contractility. PLoS One 2013; 8:e53430. [PMID: 23308222 PMCID: PMC3537614 DOI: 10.1371/journal.pone.0053430] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 11/30/2012] [Indexed: 01/17/2023] Open
Abstract
Smooth muscle contraction is a dynamic process driven by acto-myosin interactions that are controlled by multiple regulatory proteins. Our studies have shown that members of the AP-1 transcription factor family control discrete behaviors of smooth muscle cells (SMC) such as growth, migration and fibrosis. However, the role of AP-1 in regulation of smooth muscle contractility is incompletely understood. In this study we show that the AP-1 family member JunB regulates contractility in visceral SMC by altering actin polymerization and myosin light chain phosphorylation. JunB levels are robustly upregulated downstream of transforming growth factor beta-1 (TGFβ1), a known inducer of SMC contractility. RNAi-mediated silencing of JunB in primary human bladder SMC (pBSMC) inhibited cell contractility under both basal and TGFβ1-stimulated conditions, as determined using gel contraction and traction force microscopy assays. JunB knockdown did not alter expression of the contractile proteins α-SMA, calponin or SM22α. However, JunB silencing decreased levels of Rho kinase (ROCK) and myosin light chain (MLC20). Moreover, JunB silencing attenuated phosphorylation of the MLC20 regulatory phosphatase subunit MYPT1 and the actin severing protein cofilin. Consistent with these changes, cells in which JunB was knocked down showed a reduction in the F:G actin ratio in response to TGFβ1. Together these findings demonstrate a novel function for JunB in regulating visceral smooth muscle cell contractility through effects on both myosin and the actin cytoskeleton.
Collapse
Affiliation(s)
- Aruna Ramachandran
- Urological Diseases Research Center, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Samudra S. Gangopadhyay
- Urological Diseases Research Center, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ramaswamy Krishnan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sandeep A. Ranpura
- Urological Diseases Research Center, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kavitha Rajendran
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Sumati Ram-Mohan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Michelle Mulone
- Urological Diseases Research Center, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Edward M. Gong
- Urological Diseases Research Center, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rosalyn M. Adam
- Urological Diseases Research Center, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| |
Collapse
|
16
|
Suson KD, Stec AA, Shimoda LA, Gearhart JP. Initial Characterization of Exstrophy Bladder Smooth Muscle Cells in Culture. J Urol 2012; 188:1521-7. [DOI: 10.1016/j.juro.2012.02.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Indexed: 10/28/2022]
Affiliation(s)
- Kristina D. Suson
- Division of Pediatric Urology, the Brady Urological Institute, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Andrew A. Stec
- Department of Urology, Medical University of South Carolina, Charleston, South Carolina
| | - Larissa A. Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - John P. Gearhart
- Division of Pediatric Urology, the Brady Urological Institute, The Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
17
|
Di Vizio D, Morello M, Dudley AC, Schow PW, Adam RM, Morley S, Mulholland D, Rotinen M, Hager MH, Insabato L, Moses MA, Demichelis F, Lisanti MP, Wu H, Klagsbrun M, Bhowmick NA, Rubin MA, D'Souza-Schorey C, Freeman MR. Large oncosomes in human prostate cancer tissues and in the circulation of mice with metastatic disease. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:1573-84. [PMID: 23022210 DOI: 10.1016/j.ajpath.2012.07.030] [Citation(s) in RCA: 314] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 07/11/2012] [Accepted: 07/16/2012] [Indexed: 01/10/2023]
Abstract
Oncosomes are tumor-derived microvesicles that transmit signaling complexes between cell and tissue compartments. Herein, we show that amoeboid tumor cells export large (1- to 10-μm diameter) vesicles, derived from bulky cellular protrusions, that contain metalloproteinases, RNA, caveolin-1, and the GTPase ADP-ribosylation factor 6, and are biologically active toward tumor cells, endothelial cells, and fibroblasts. We describe methods by which large oncosomes can be selectively sorted by flow cytometry and analyzed independently of vesicles <1 μm. Structures resembling large oncosomes were identified in the circulation of different mouse models of prostate cancer, and their abundance correlated with tumor progression. Similar large vesicles were also identified in human tumor tissues, but they were not detected in the benign compartment. They were more abundant in metastases. Our results suggest that tumor microvesicles substantially larger than exosome-sized particles can be visualized and quantified in tissues and in the circulation, and isolated and characterized using clinically adaptable methods. These findings also suggest a mechanism by which migrating tumor cells condition the tumor microenvironment and distant sites, thereby potentiating advanced disease.
Collapse
Affiliation(s)
- Dolores Di Vizio
- Division of Cancer Biology and Therapeutics, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Wang Z, Cheng Z, Cristofaro V, Li J, Xiao X, Gomez P, Ge R, Gong E, Strle K, Sullivan MP, Adam RM, White MF, Olumi AF. Inhibition of TNF-α improves the bladder dysfunction that is associated with type 2 diabetes. Diabetes 2012; 61:2134-45. [PMID: 22688336 PMCID: PMC3402324 DOI: 10.2337/db11-1763] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Diabetic bladder dysfunction (DBD) is common and affects 80% of diabetic patients. However, the molecular mechanisms underlying DBD remain elusive because of a lack of appropriate animal models. We demonstrate DBD in a mouse model that harbors hepatic-specific insulin receptor substrate 1 and 2 deletions (double knockout [DKO]), which develops type 2 diabetes. Bladders of DKO animals exhibited detrusor overactivity at an early stage: increased frequency of nonvoiding contractions during bladder filling, decreased voided volume, and dispersed urine spot patterns. In contrast, older animals with diabetes exhibited detrusor hypoactivity, findings consistent with clinical features of diabetes in humans. The tumor necrosis factor (TNF) superfamily genes were upregulated in DKO bladders. In particular, TNF-α was upregulated in serum and in bladder smooth muscle tissue. TNF-α augmented the contraction of primary cultured bladder smooth muscle cells through upregulating Rho kinase activity and phosphorylating myosin light chain. Systemic treatment of DKO animals with soluble TNF receptor 1 (TNFRI) prevented upregulation of Rho A signaling and reversed the bladder dysfunction, without affecting hyperglycemia. TNFRI combined with the antidiabetic agent, metformin, improved DBD beyond that achieved with metformin alone, suggesting that therapies targeting TNF-α may have utility in reversing the secondary urologic complications of type 2 diabetes.
Collapse
Affiliation(s)
- Zongwei Wang
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Zhiyong Cheng
- Division of Endocrinology, Howard Hughes Medical Institute, Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Vivian Cristofaro
- Urology Research, Veterans Administration Boston Healthcare System, Harvard Medical School, Boston, Massachusetts
| | - Jijun Li
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Integrative Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xingyuan Xiao
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Urology, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Pablo Gomez
- Urology Research Center, Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Rongbin Ge
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Edward Gong
- Urology Research Center, Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Klemen Strle
- Department of Medicine, Division of Allergy/Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Maryrose P. Sullivan
- Urology Research, Veterans Administration Boston Healthcare System, Harvard Medical School, Boston, Massachusetts
| | - Rosalyn M. Adam
- Urology Research Center, Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Morris F. White
- Division of Endocrinology, Howard Hughes Medical Institute, Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Aria F. Olumi
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Corresponding author: Aria F. Olumi,
| |
Collapse
|
19
|
Monaghan KP, Johnston L, McCloskey KD. Identification of PDGFRα positive populations of interstitial cells in human and guinea pig bladders. J Urol 2012; 188:639-47. [PMID: 22704452 DOI: 10.1016/j.juro.2012.03.117] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Indexed: 02/02/2023]
Abstract
PURPOSE The bladder wall comprises a complex array of cells, including urothelium, smooth muscle, nerves and interstitial cells. Interstitial cells have several subtypes based on site, morphology and differential expression of markers such as anti-vimentin and anti-KIT. We examined whether a subpopulation of interstitial cells immunopositive for PDGFRα exists in human and guinea pig bladders. MATERIALS AND METHODS Human and guinea pig bladder tissues were processed for immunohistochemistry and examined by bright field or confocal microscopy. Whole mount tissues and paraffin sections were labeled with antibodies to PDGFRα, vimentin, KIT and PGP9.5. Protein expression was assessed by Western blot. RESULTS PDGFRα(+) cells were present in human and guinea pig bladders. In the guinea pig PDGFRα(+) cells had a branched stellate morphology and formed networks in the lamina propria. In human and guinea pig detrusors PDGFRα(+) cells were elongated on the boundary of smooth muscle bundles or were seen as groups of stellate cells in the interbundle spaces. PDGFRα(+) cells were located close to nerves labeled by PGP9.5. Double labeling revealed that PDGFRα(+) cells were a subgroup of the vimentin(+) population. A significant proportion of PDGFRα(+) cells were also KIT(+). Bands corresponding to PDGFRα, KIT and vimentin proteins were detected on Western blot. CONCLUSIONS To our knowledge this study is the first to identify PDGFRα(+)/KIT(+) cells in the bladder lamina propria and detrusor layers. These cells are a subgroup of the vimentin(+) population, showing the complexity of bladder interstitial cells. PDGFRα(+) cells are apparently structurally associated with intramural nerves, indicating integration with bladder control mechanisms.
Collapse
Affiliation(s)
- Kevin P Monaghan
- Centre for Cancer Research and Cell Biology, Queen's University, Belfast, United Kingdom
| | | | | |
Collapse
|
20
|
Increased smooth muscle contractility in mice deficient for neuropilin 2. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:548-59. [PMID: 22688055 DOI: 10.1016/j.ajpath.2012.04.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 03/30/2012] [Accepted: 04/05/2012] [Indexed: 12/28/2022]
Abstract
Neuropilins (NRPs) are transmembrane receptors that bind class 3 semaphorins and VEGF family members to regulate axon guidance and angiogenesis. Although expression of NRP1 by vascular smooth muscle cells (SMCs) has been reported, NRP function in smooth muscle (SM) in vivo is unexplored. Using Nrp2(+/LacZ) and Nrp2(+/gfp) transgenic mice, we observed robust and sustained expression of Nrp2 in the SM compartments of the bladder and gut, but no expression in vascular SM, skeletal muscle, or cardiac muscle. This expression pattern was recapitulated in vitro using primary human SM cell lines. Alterations in cell morphology after treatment of primary visceral SMCs with the NRP2 ligand semaphorin-3F (SEMA3F) were accompanied by inhibition of RhoA activity and myosin light chain phosphorylation, as well as decreased cytoskeletal stiffness. Ex vivo contractility testing of bladder muscle strips exposed to electrical stimulation or soluble agonists revealed enhanced tension generation of tissues from mice with constitutive or SM-specific knockout of Nrp2, compared with controls. Mice lacking Nrp2 also displayed increased bladder filling pressures, as assessed by cystometry in conscious mice. Together, these findings identify Nrp2 as a mediator of prorelaxant stimuli in SMCs and suggest a novel function for Nrp2 as a regulator of visceral SM contractility.
Collapse
|
21
|
Ramachandran A, Gong EM, Pelton K, Ranpura SA, Mulone M, Seth A, Gomez P, Adam RM. FosB regulates stretch-induced expression of extracellular matrix proteins in smooth muscle. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:2977-89. [PMID: 21996678 DOI: 10.1016/j.ajpath.2011.08.034] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2011] [Revised: 07/29/2011] [Accepted: 08/09/2011] [Indexed: 11/19/2022]
Abstract
Fibroproliferative remodeling in smooth muscle-rich hollow organs is associated with aberrant extracellular matrix (ECM) production. Although mechanical stimuli regulate ECM protein expression, the transcriptional mediators of this process remain poorly defined. Previously, we implicated AP-1 as a mediator of smooth muscle cell (SMC) mechanotransduction; however, its role in stretch-induced ECM regulation has not been explored. Herein, we identify a novel role for the AP-1 subunit FosB in stretch-induced ECM expression in SMCs. The DNA-binding activity of AP-1 increased after stretch stimulation of SMCs in vitro. In contrast to c-Jun and c-fos, which are also activated by the SMC mitogen platelet-derived growth factor, FosB was only activated by stretch. FosB silencing attenuated the expression of the profibrotic factors tenascin C (TNC) and connective tissue growth factor (CTGF), whereas forced expression of Jun~FosB stimulated TNC and CTGF promoter activity. Chromatin immunoprecipitation revealed enrichment of AP-1 at the TNC and CTGF promoters. Bladder distension in vivo enhanced nuclear localization of c-jun and FosB. Finally, the distension-induced expression of TNC and CTGF in the detrusor smooth muscle of bladders from wild-type mice was significantly attenuated in FosB-null mice. Together, these findings identify FosB as a mechanosensitive regulator of ECM production in smooth muscle.
Collapse
Affiliation(s)
- Aruna Ramachandran
- Urological Diseases Research Center, Children's Hospital Boston, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Remodeling of uterine spiral arteries by trophoblast cells is a requisite process for hemochorial placentation and successful pregnancy. The rat exhibits deep intrauterine trophoblast invasion and accompanying trophoblast-directed vascular modification. The involvement of phosphatidylinositol 3 kinase (PI3K), AKT, and Fos-like antigen 1 (FOSL1) in regulating invasive trophoblast and hemochorial placentation was investigated using Rcho-1 trophoblast stem cells and rat models. Disruption of PI3K/AKT with small-molecule inhibitors interfered with the differentiation-dependent elaboration of a signature invasive-vascular remodeling trophoblast gene expression profile and trophoblast invasion. AKT isoform-specific knockdown also affected the signature invasive-vascular remodeling trophoblast gene expression profile. Nuclear FOSL1 increased during trophoblast cell differentiation in a PI3K/AKT-dependent manner. Knockdown of FOSL1 disrupted the expression of a subset of genes associated with the invasive-vascular remodeling trophoblast phenotype, including the matrix metallopeptidase 9 gene (Mmp9). FOSL1 was shown to occupy regions of the Mmp9 promoter in trophoblast cells critical for the regulation of Mmp9 gene expression. Inhibition of FOSL1 expression also abrogated trophoblast invasion, as assessed in vitro and following in vivo trophoblast-specific lentivirally delivered FOSL1 short hairpin RNA (shRNA). In summary, FOSL1 is a key downstream effector of the PI3K/AKT signaling pathway responsible for development of trophoblast lineages integral to establishing the maternal-fetal interface.
Collapse
|