1
|
Ameer G, Keate R, Bury M, Mendez-Santos M, Gerena A, Goedegebuure M, Rivnay J, Sharma A. Cell-free biodegradable electroactive scaffold for urinary bladder regeneration. RESEARCH SQUARE 2024:rs.3.rs-3817836. [PMID: 38352487 PMCID: PMC10862962 DOI: 10.21203/rs.3.rs-3817836/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
Tissue engineering heavily relies on cell-seeded scaffolds to support the complex biological and mechanical requirements of a target organ. However, in addition to safety and efficacy, translation of tissue engineering technology will depend on manufacturability, affordability, and ease of adoption. Therefore, there is a need to develop scalable biomaterial scaffolds with sufficient bioactivity to eliminate the need for exogenous cell seeding. Herein, we describe synthesis, characterization, and implementation of an electroactive biodegradable elastomer for urinary bladder tissue engineering. To create an electrically conductive and mechanically robust scaffold to support bladder tissue regeneration, we developed a phase-compatible functionalization method wherein the hydrophobic conductive polymer poly(3,4-ethylenedioxythiophene) (PEDOT) was polymerized in situ within a similarly hydrophobic citrate-based elastomer poly(octamethylene-citrate-co-octanol) (POCO) film. We demonstrate the efficacy of this film as a scaffold for bladder augmentation in athymic rats, comparing PEDOT-POCO scaffolds to mesenchymal stromal cell-seeded POCO scaffolds. PEDOT-POCO recovered bladder function and anatomical structure comparably to the cell-seeded POCO scaffolds and significantly better than non-cell seeded POCO scaffolds. This manuscript reports: (1) a new phase-compatible functionalization method that confers electroactivity to a biodegradable elastic scaffold, and (2) the successful restoration of the anatomy and function of an organ using a cell-free electroactive scaffold.
Collapse
|
2
|
Yang B, Yang G, Zhao F, Yao X, Xu L, Zhou L. Autologous Endothelial Progenitor Cells and Bioactive Factors Improve Bladder Regeneration. Tissue Eng Part C Methods 2024; 30:15-26. [PMID: 37756374 DOI: 10.1089/ten.tec.2023.0079] [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] [Indexed: 09/29/2023] Open
Abstract
Insufficient vascularization is still a challenge that impedes bladder tissue engineering and results in unsatisfied smooth muscle regeneration. Since bladder regeneration is a complex articulated process, the aim of this study is to investigate whether combining multiple pathways by exploiting a combination of biomaterials, cells, and bioactive factors, contributes to the improvements of smooth muscle regeneration and vascularization in tissue-engineered bladder. Autologous endothelial progenitor cells (EPCs) and bladder smooth muscle cells (BSMCs) are cultured and incorporated into our previously prepared porcine bladder acellular matrix (BAM) for bladder augmentation in rabbits. Simultaneously, exogenous vascular endothelial growth factor (VEGF) and platelet-derived growth factor BB (PDGF-BB) mixed with Matrigel were injected around the implanted cells-BAM complex. In the results, compared with control rabbits received bladder augmentation with porcine BAM seeded with BSMCs, the experimental animals showed significantly improved smooth muscle regeneration and vascularization, along with more excellent functional recovery of tissue-engineered bladder, due to the additional combination of autologous EPCs and bioactive factors, including VEGF and PDGF-BB. Furthermore, cell tracking suggested that the seeded EPCs could be directly involved in neovascularization. Therefore, it may be an effective method to combine multiple pathways for tissue-engineering urinary bladder.
Collapse
Affiliation(s)
- Bin Yang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guanjie Yang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Feng Zhao
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Luwei Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Liuhua Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
3
|
The Potential of Cell Sheet Technology for Beta Cell Replacement Therapy. CURRENT TRANSPLANTATION REPORTS 2022. [DOI: 10.1007/s40472-022-00371-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Abstract
Purpose of Review
Here, we review the use of cell sheet technology using different cell types and its potential for restoring the extracellular matrix microenvironment, perfusion, and immunomodulatory action on islets and beta cells.
Recent Findings
Cell sheets can be produced with different fabrication techniques ranging from the widely used temperature responsive system to the magnetic system. A variety of cells have been used to produce cell sheets including skin fibroblasts, smooth muscle cells, human umbilical vein endothelial cells, and mesenchymal stem cells.
Summary
CST would allow to recreate the ECM of islets which would provide cues to support islet survival and improvement of islet function. Depending on the used cell type, different additional supporting properties like immunoprotection or cues for better revascularization could be provided. Furthermore, CST offers the possibility to use other implantation sites than inside the liver. Further research should focus on cell sheet thickness and size to generate a potential translational therapy.
Collapse
|
4
|
Zhang W, Hu J, Huang Y, Wu C, Xie H. Urine-derived stem cells: applications in skin, bone and articular cartilage repair. BURNS & TRAUMA 2021; 9:tkab039. [PMID: 34859109 PMCID: PMC8633594 DOI: 10.1093/burnst/tkab039] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/18/2021] [Indexed: 02/05/2023]
Abstract
As an emerging type of adult stem cell featuring non-invasive acquisition, urine-derived stem cells (USCs) have shown great potential for applications in tissue engineering and regenerative medicine. With a growing amount of research on the topic, the effectiveness of USCs in various disease models has been shown and the underlying mechanisms have also been explored, though many aspects still remain unclear. In this review, we aim to provide an up-to-date overview of the biological characteristics of USCs and their applications in skin, bone and articular cartilage repair. In addition to the identification procedure of USCs, we also summarize current knowledge of the underlying repair mechanisms and application modes of USCs. Potential concerns and perspectives have also been summarized.
Collapse
Affiliation(s)
- Wenqian Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jungen Hu
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yizhou Huang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chenyu Wu
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Huiqi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
5
|
Kurdi BA, Ababneh NA, Abuharfeil N, Al Demour S, Awidi AS. Use of conditioned media (CM) and xeno-free serum substitute on human adipose-derived stem cells (ADSCs) differentiation into urothelial-like cells. PeerJ 2021; 9:e10890. [PMID: 33850639 PMCID: PMC8019311 DOI: 10.7717/peerj.10890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/12/2021] [Indexed: 01/01/2023] Open
Abstract
Background Congenital abnormalities, cancers as well as injuries can cause irreversible damage to the urinary tract, which eventually requires tissue reconstruction. Smooth muscle cells, endothelial cells, and urothelial cells are the major cell types required for the reconstruction of lower urinary tract. Adult stem cells represent an accessible source of unlimited repertoire of untransformed cells. Aim Fetal bovine serum (FBS) is the most vital supplement in the culture media used for cellular proliferation and differentiation. However, due to the increasing interest in manufacturing xeno-free stem cell-based cellular products, optimizing the composition of the culture media and the serum-type used is of paramount importance. In this study, the effects of FBS and pooled human platelet (pHPL) lysate were assessed on the capacity of human adipose-derived stem cells (ADSCs) to differentiate into urothelial-like cells. Also, we aimed to compare the ability of both conditioned media (CM) and unconditioned urothelial cell media (UCM) to induce urothelial differentiation of ADCS in vitro. Methods ADSCs were isolated from human lipoaspirates and characterized by flow cytometry for their ability to express the most common mesenchymal stem cell (MSCs) markers. The differentiation potential was also assessed by differentiating them into osteogenic and adipogenic cell lineages. To evaluate the capacity of ADSCs to differentiate towards the urothelial-like lineage, cells were cultured with either CM or UCM, supplemented with either 5% pHPL, 2.5% pHPL or 10% FBS. After 14 days of induction, cells were utilized for gene expression and immunofluorescence analysis. Results ADSCs cultured in CM and supplemented with FBS exhibited the highest upregulation levels of the urothelial cell markers; cytokeratin-18 (CK-18), cytokeratin-19 (CK-19), and Uroplakin-2 (UPK-2), with a 6.7, 4.2- and a 2-folds increase in gene expression, respectively. Meanwhile, the use of CM supplemented with either 5% pHPL or 2.5% pHPL, and UCM supplemented with either 5% pHPL or 2.5% pHPL showed low expression levels of CK-18 and CK-19 and no upregulation of UPK-2 level was observed. In contrast, the use of UCM with FBS has increased the levels of CK-18 and CK-19, however to a lesser extent compared to CM. At the cellular level, CK-18 and UPK-2 were only detected in CM/FBS supplemented group. Growth factor analysis revealed an increase in the expression levels of EGF, VEGF and PDGF in all of the differentiated groups. Conclusion Efficient ADSCs urothelial differentiation is dependent on the use of conditioned media. The presence of high concentrations of proliferation-inducing growth factors present in the pHPL reduces the efficiency of ADSCs differentiation towards the urothelial lineage. Additionally, the increase in EGF, VEGF and PDGF during the differentiation implicates them in the mechanism of urothelial cell differentiation.
Collapse
Affiliation(s)
- Ban Al- Kurdi
- Cell Therapy Center, University of Jordan, Amman, Jordan.,Department of Hematology and Oncology, Jordan University Hospital, Amman, Jordan
| | | | - Nizar Abuharfeil
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, Jordan
| | - Saddam Al Demour
- Department of Urology, School of medicine, University of Jordan, Amman, Jordan, University of Jordan, Amman, Jordan
| | - Abdalla S Awidi
- Cell Therapy Center, University of Jordan, Amman, Jordan.,Department of Hematology and Oncology, Jordan University Hospital, Amman, Jordan
| |
Collapse
|
6
|
Effect of PDGF-B Gene-Activated Acellular Matrix and Mesenchymal Stem Cell Transplantation on Full Thickness Skin Burn Wound in Rat Model. Tissue Eng Regen Med 2020; 18:235-251. [PMID: 33145744 DOI: 10.1007/s13770-020-00302-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 08/10/2020] [Accepted: 09/16/2020] [Indexed: 10/23/2022] Open
Abstract
BACKGROUND Full thickness burn wounds are lack of angiogenesis, cell migration, epithelialisation and finally scar tissue formation. Tissue engineered composite graft can provide sustained release of growth factor and promote the wound healing by cell migration, early angiogenesis and proliferation of extracellular matrix and wound remodeling. The objective of this study was to evaluate the gene embedded (pDNA-platelet-derived growth factor, PDGF-B) porcine acellular urinary bladder matrix with transfected mesenchymal stem cells (rBMSC) on healing of full thickness burn wound in rat model. METHODS Full thickness burn wound of 2 × 2 cm size was created in dorsum of rat model under general anesthesia. Burn wounds were treated with silver sulfadiazine; porcine acellular urinary bladder matrix (PAUBM); PAUBM transfected with pDNA-PDGF-B; PAUBM seeded with rBMSC; PAUBM seeded with rBMSC transfected with pDNA-PDGF-B in groups A, B, C, D and E respectively. The wound healing was assessed based on clinical, macroscopically, immunologically, histopathological and RT-qPCR parameters. RESULTS Wound was significantly healed in group E and group D with early extracellular matrix deposition, enhanced granulation tissue formation and early angiogenesis compared to all other groups. The immunologic response against porcine acellular matrix showed that PDGF-B gene activated matrix along with stem cell group showed less antibody titer against acellular matrix than other groups in all intervals. PDGF gene activated matrix releasing the PDGF-B and promote the healing of full thickness burn wound with neovascularization and neo tissue formation. PDGF gene also enhances secretion of other growth factors results in PDGF mediated regenerative activities. This was confirmed in RT-qPCR at various time intervals. CONCLUSION Gene activated matrix encoded for PDGF-B protein transfected stem cells have been clinically proven for early acceleration of angiogenesis and tissue regeneration in burn wounds in rat models. Evaluation of PDGF-B gene-activated acellular matrix and mesenchymal stem cell in full thickness skin burn wound in rat.
Collapse
|
7
|
Yu Y, Jiang W. Pluripotent stem cell differentiation as an emerging model to study human prostate development. Stem Cell Res Ther 2020; 11:285. [PMID: 32678004 PMCID: PMC7364497 DOI: 10.1186/s13287-020-01801-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/18/2020] [Accepted: 07/01/2020] [Indexed: 12/11/2022] Open
Abstract
Prostate development is a complex process, and knowledge about this process is increasingly required for both basic developmental biology studies and clinical prostate cancer research, as prostate tumorigenesis can be regarded as the restoration of development in the adult prostate. Using rodent animal models, scientists have revealed that the development of the prostate is mainly mediated by androgen receptor (AR) signaling and that some other signaling pathways also play indispensable roles. However, there are still many unknowns in human prostate biology, mainly due to the limited availability of proper fetal materials. Here, we first briefly review prostate development with a focus on the AR, WNT, and BMP signaling pathways is necessary for prostate budding/BMP signaling pathways. Based on the current progress in in vitro prostatic differentiation and organoid techniques, we propose human pluripotent stem cells as an emerging model to study human prostate development.
Collapse
Affiliation(s)
- Yangyang Yu
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, 116 East-Lake Road, District of Wuchang, Wuhan, 430071, Hubei Province, China
| | - Wei Jiang
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, 116 East-Lake Road, District of Wuchang, Wuhan, 430071, Hubei Province, China. .,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China. .,Human Genetics Resource Preservation Center of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
8
|
Osei-Amponsa V, Buckwalter JM, Shuman L, Zheng Z, Yamashita H, Walter V, Wildermuth T, Ellis-Mohl J, Liu C, Warrick JI, Shantz LM, Feehan RP, Al-Ahmadie H, Mendelsohn C, Raman JD, Kaestner KH, Wu XR, DeGraff DJ. Hypermethylation of FOXA1 and allelic loss of PTEN drive squamous differentiation and promote heterogeneity in bladder cancer. Oncogene 2019; 39:1302-1317. [PMID: 31636388 DOI: 10.1038/s41388-019-1063-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 10/02/2019] [Accepted: 10/07/2019] [Indexed: 01/04/2023]
Abstract
Intratumoral heterogeneity in bladder cancer is a barrier to accurate molecular sub-classification and treatment efficacy. However, individual cellular and mechanistic contributions to tumor heterogeneity are controversial. We examined potential mechanisms of FOXA1 and PTEN inactivation in bladder cancer and their contribution to tumor heterogeneity. These analyses were complemented with inactivation of FOXA1 and PTEN in intermediate and luminal mouse urothelium. We show inactivation and reduced expression of FOXA1 and PTEN is prevalent in human disease, where PTEN and FOXA1 are downregulated by allelic loss and site-specific DNA hypermethylation, respectively. Conditional inactivation of both Foxa1 and Pten in intermediate/luminal cells in mice results in development of bladder cancer exhibiting squamous features as well as enhanced sensitivity to a bladder-specific carcinogen. In addition, FOXA1 is hypermethylated in basal bladder cancer cell lines, and this is reversed by treatment with DNA methyltransferase inhibitors. By integrating human correlative and in vivo studies, we define a critical role for PTEN loss and epigenetic silencing of FOXA1 in heterogeneous human disease and show genetic targeting of luminal/intermediate cells in mice drives squamous differentiation.
Collapse
Affiliation(s)
- Vasty Osei-Amponsa
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Jenna M Buckwalter
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.,Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Lauren Shuman
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.,Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Zongyu Zheng
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Hironobu Yamashita
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.,Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Vonn Walter
- Department of Public Health Sciences, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Thomas Wildermuth
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.,Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Justine Ellis-Mohl
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.,Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Chang Liu
- Department of Urology, Columbia University, New York, NY, USA
| | - Joshua I Warrick
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.,Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Lisa M Shantz
- Department of Cellular and Molecular Physiology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Robert P Feehan
- Department of Cellular and Molecular Physiology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Hikmat Al-Ahmadie
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Jay D Raman
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.,Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Klaus H Kaestner
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Xue-Ru Wu
- Departments of Urology and Pathology, New York University School of Medicine, New York, NY, USA.,Veterans Affairs New York Harbor Healthcare System, Manhattan Campus, New York, NY, USA
| | - David J DeGraff
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA. .,Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.
| |
Collapse
|
9
|
Zhou L, Xia J, Wang P, Jia R, Zheng J, Yao X, Chen Y, Dai Y, Yang B. Autologous Smooth Muscle Progenitor Cells Enhance Regeneration of Tissue-Engineered Bladder. Tissue Eng Part A 2018; 24:1066-1081. [PMID: 29327677 DOI: 10.1089/ten.tea.2017.0376] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Liuhua Zhou
- Department of Urology and Andrology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, China
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jiadong Xia
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Pengji Wang
- Department of Urology and Andrology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, China
- Department of Urology, Longkou People Hospital, Yantai, China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Junhua Zheng
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yun Chen
- Department of Urology and Andrology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yutian Dai
- Department of Urology and Andrology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Bin Yang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Bioengineering Approaches for Bladder Regeneration. Int J Mol Sci 2018; 19:ijms19061796. [PMID: 29914213 PMCID: PMC6032229 DOI: 10.3390/ijms19061796] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/06/2018] [Accepted: 06/10/2018] [Indexed: 12/25/2022] Open
Abstract
Current clinical strategies for bladder reconstruction or substitution are associated to serious problems. Therefore, new alternative approaches are becoming more and more necessary. The purpose of this work is to review the state of the art of the current bioengineering advances and obstacles reported in bladder regeneration. Tissue bladder engineering requires an ideal engineered bladder scaffold composed of a biocompatible material suitable to sustain the mechanical forces necessary for bladder filling and emptying. In addition, an engineered bladder needs to reconstruct a compliant muscular wall and a highly specialized urothelium, well-orchestrated under control of autonomic and sensory innervations. Bioreactors play a very important role allowing cell growth and specialization into a tissue-engineered vascular construct within a physiological environment. Bioprinting technology is rapidly progressing, achieving the generation of custom-made structural supports using an increasing number of different polymers as ink with a high capacity of reproducibility. Although many promising results have been achieved, few of them have been tested with clinical success. This lack of satisfactory applications is a good reason to discourage researchers in this field and explains, somehow, the limited high-impact scientific production in this area during the last decade, emphasizing that still much more progress is required before bioengineered bladders become a commonplace in the clinical setting.
Collapse
|
11
|
Osborn SL, Kurzrock EA. In Vitro Differentiation and Propagation of Urothelium from Pluripotent Stem Cell Lines. Methods Mol Biol 2018; 1655:137-144. [PMID: 28889383 DOI: 10.1007/978-1-4939-7234-0_11] [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] [Indexed: 01/16/2023]
Abstract
Bioengineering of bladder tissue, particularly for those patients who have advanced bladder disease, requires a source of urothelium that is healthy, capable of significant proliferation in vitro and immunologically tolerated upon transplant. As pluripotent stem cells have the potential to fulfill such criteria, they provide a critical cell source from which urothelium might be derived in vitro and used clinically. Herein, we describe the in vitro differentiation of urothelium from the H9 human embryonic stem cell (hESC) line through the definitive endoderm (DE) phase via selective culture techniques. The protocol can be used to derive urothelium from other hESCs or human-induced pluripotent stem cells.
Collapse
Affiliation(s)
- Stephanie L Osborn
- Department of Urology, University of California, Davis School of Medicine, 4860 Y Street, Suite 3500, Sacramento, CA, 95817, USA.,Stem Cell Program, Institute for Regenerative Cures, University of California, Davis Medical Center, Sacramento, CA, 95817, USA
| | - Eric A Kurzrock
- Department of Urology, University of California, Davis School of Medicine, 4860 Y Street, Suite 3500, Sacramento, CA, 95817, USA. .,Stem Cell Program, Institute for Regenerative Cures, University of California, Davis Medical Center, Sacramento, CA, 95817, USA.
| |
Collapse
|
12
|
Huang CP, Chen CC, Shyr CR. The anti-tumor effect of intravesical administration of normal urothelial cells on bladder cancer. Cytotherapy 2017; 19:1233-1245. [PMID: 28818454 DOI: 10.1016/j.jcyt.2017.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 06/12/2017] [Accepted: 06/25/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND AIMS Urothelial bladder cancer (UBC) is the second most common cancer of the genitourinary tract and for advanced forms of the disease it has a high mortality rate. There are no approved new molecularly targeted agents or chemotherapeutics for advanced UBC beyond cisplatin-based chemotherapy except the recently approved anti-programmed death ligand 1 (anti-PD-1/PD-L1) antibody. With complex genetic and epigenetic alterations in tumors, despite several druggable targets identified, to cure UBC is still a challenging unmet medical need. Like other cancers, UBC to the host body is considered as a wound, aging stem cell disease and immunosuppressive disorder. Therefore, we proposed a novel cellular approach to target the host body by intravesical instilling of normal urothelial cells that could repair the injury and reduce inflammation by activating body-reparative capacity and because non-self cells are transplanted, host body immune responses could be induced in the tumor microenvironment of UBC to restrain and even eliminate tumor cells. METHODS In this study, we isolated and expanded normal male murine urothelial cells and intravesically administered them into the bladders of female mice of two orthotopic bladder tumor models and one urothelial injury model. RESULTS We showed that the instillation of normal urothelial cells containing stem/progenitor cell population into bladders could have anti-tumor effect in orthotopic tumor models, possibly by activating immune responses and helping injured urothelium tissue recovery in a chemically induced urothelial injury model. CONCLUSIONS Our findings could lead to an innovative and revolutionary cell therapy modality with normal urothelial cells as an effective and safe therapeutic option for UBC.
Collapse
Affiliation(s)
- Chi-Ping Huang
- Sex Hormone Research Center, Departments of Urology/Surgery and Medical Laboratory Science & Biotechnology, Graduate Institute of Clinical Medical Science, China Medical University/Hospital, Taichung, Taiwan
| | - Chi-Cheng Chen
- Sex Hormone Research Center, Departments of Urology/Surgery and Medical Laboratory Science & Biotechnology, Graduate Institute of Clinical Medical Science, China Medical University/Hospital, Taichung, Taiwan; Department of Urology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan
| | - Chih-Rong Shyr
- Sex Hormone Research Center, Departments of Urology/Surgery and Medical Laboratory Science & Biotechnology, Graduate Institute of Clinical Medical Science, China Medical University/Hospital, Taichung, Taiwan.
| |
Collapse
|
13
|
Al-Kurdi B. Hierarchical transcriptional profile of urothelial cells development and differentiation. Differentiation 2017; 95:10-20. [PMID: 28135607 DOI: 10.1016/j.diff.2016.10.001] [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: 04/30/2016] [Revised: 10/09/2016] [Accepted: 10/14/2016] [Indexed: 11/27/2022]
Abstract
The urothelial lining of the lower urinary tract is the most efficient permeability barrier in animals, exhibiting a highly differentiated phenotype and a remarkable regenerative capacity upon wounding. During development and possibly during repair, cells undergo a sequence of hierarchical transcriptional events that mark the transition of these cells from the least differentiated urothelial phenotype characteristic of the basal cell layer, to the most differentiated cellular phenotype characteristic of the superficial cell layer. Unraveling normal urothelial differentiation program is essential to uncover the underlying causes of many congenital abnormalities and for the development of an appropriate differentiation niche for stem cells, for future use in urinary tract tissue engineering and organ reconstruction. Kruppel like factor-5 appears to be at the top of the hierarchy activating several downstream transcription factors, the most prominent of which is peroxisome proliferator activator receptor-γ. Eventually those lead to the activation of transcription factors that directly regulate the expression of uroplakin proteins along with other proteins that mediate the permeability function of the urothelium. In this review, we discuss the most recent findings in the area of urothelial cellular differentiation and transcriptional regulation, aiming for a comprehensive overview that aids in a refined understanding of this process.
Collapse
Affiliation(s)
- Ban Al-Kurdi
- Cell Therapy Center, The University of Jordan, Amman, Jordan.
| |
Collapse
|
14
|
de Graaf P, van der Linde EM, Rosier PFWM, Izeta A, Sievert KD, Bosch JLHR, de Kort LMO. Systematic Review to Compare Urothelium Differentiation with Urethral Epithelium Differentiation in Fetal Development, as a Basis for Tissue Engineering of the Male Urethra. TISSUE ENGINEERING PART B-REVIEWS 2016; 23:257-267. [PMID: 27809709 DOI: 10.1089/ten.teb.2016.0352] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Tissue-engineered (TE) urethra is desirable in men with urethral disease (stricture or hypospadias) and shortage of local tissue. Although ideally a TE graft would contain urethral epithelium cells, currently, bladder epithelium (urothelium) is widely used, but morphologically different. Understanding the differences and similarities of urothelium and urethral epithelium could help design a protocol for in vitro generation of urethral epithelium to be used in TE grafts for the urethra. PURPOSE To understand the development toward urethral epithelium or urothelium to improve TE of the urethra. METHODS A literature search was done following PRISMA guidelines. Articles describing urethral epithelium and bladder urothelium development in laboratory animals and humans were selected. RESULTS Twenty-nine studies on development of urethral epithelium and 29 studies on development of urothelium were included. Both tissue linings derive from endoderm and although adult urothelium and urethral epithelium are characterized by different gene expression profiles, the signaling pathways underlying their development are similar, including Shh, BMP, Wnt, and FGF. The progenitor of the urothelium and the urethral epithelium is the early fetal urogenital sinus (UGS). The urethral plate and the urothelium are both formed from the p63+ cells of the UGS. Keratin 20 and uroplakins are exclusively expressed in urothelium, not in the urethral epithelium. Further research has to be done on unique markers for the urethral epithelium. CONCLUSION This review has summarized the current knowledge about embryonic development of urothelium versus urethral epithelium and especially focuses on the influencing factors that are potentially specific for the eventual morphological differences of both cell linings, to be a basis for developmental or tissue engineering of urethral tissue.
Collapse
Affiliation(s)
- Petra de Graaf
- 1 Department of Urology, University Medical Centre Utrecht , Utrecht, The Netherlands .,2 Regenerative Medicine Center Utrecht , Utrecht, The Netherlands
| | | | - Peter F W M Rosier
- 1 Department of Urology, University Medical Centre Utrecht , Utrecht, The Netherlands
| | - Ander Izeta
- 3 Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, Hospital Universitario Donostia , San Sebastián, Spain .,4 Department of Biomedical Engineering, School of Engineering, Tecnun-University of Navarra , San Sebastián, Spain
| | | | - J L H Ruud Bosch
- 1 Department of Urology, University Medical Centre Utrecht , Utrecht, The Netherlands
| | - Laetitia M O de Kort
- 1 Department of Urology, University Medical Centre Utrecht , Utrecht, The Netherlands
| |
Collapse
|
15
|
Moschouris K, Firoozi N, Kang Y. The application of cell sheet engineering in the vascularization of tissue regeneration. Regen Med 2016; 11:559-70. [PMID: 27527673 PMCID: PMC5007660 DOI: 10.2217/rme-2016-0059] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Scaffold-free cell sheet engineering (CSE) is a new technology to regenerate injured or damaged tissues, which has shown promising potential in tissue regeneration. CSE uses a thermosensitive surface to form a dense cell sheet that can be detached when temperature decreases. The detached cell sheet can be stacked on top of one another according to the thickness of cell sheet for the specific tissue regeneration application. One of the key challenges of tissue engineering is vascularization. CSE technique provides excellent microenvironment for vascularization since the technique can maintain the intact cell matrix that is crucial for angiogenesis. In this review paper, we will highlight the principle technique of CSE and its application in tissue regeneration.
Collapse
Affiliation(s)
- Kathryn Moschouris
- Department of Biological Sciences, College of Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Negar Firoozi
- Department of Ocean & Mechanical Engineering, College of Engineering & Computer Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Yunqing Kang
- Department of Ocean & Mechanical Engineering, College of Engineering & Computer Science, Florida Atlantic University, Boca Raton, FL 33431, USA.,Department of Biomedical Science, College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| |
Collapse
|
16
|
Bunimovich-Mendrazitsky S, Pisarev V, Kashdan E. Modeling and simulation of a low-grade urinary bladder carcinoma. Comput Biol Med 2015; 58:118-29. [DOI: 10.1016/j.compbiomed.2014.12.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 12/26/2014] [Indexed: 10/24/2022]
|
17
|
Anabolic androgens affect the competitive interactions in cell migration and adhesion between normal mouse urothelial cells and urothelial carcinoma cells. Biochem Biophys Res Commun 2014; 452:322-7. [PMID: 25159849 DOI: 10.1016/j.bbrc.2014.08.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 08/08/2014] [Indexed: 12/17/2022]
Abstract
The urothelium is constantly rebuilt by normal urothelial cells to regenerate damaged tissues caused by stimuli in urine. However, the urothelial carcinoma cells expand the territory by aberrant growth of tumor cells, which migrate and occupy the damaged tissues to spread outside and disrupt the normal cells and organized tissues and form a tumor. Therefore, the interaction between normal urothelial cells and urothelial carcinoma cells affect the initiation and progression of urothelial tumors if normal urothelial cells fail to migrate and adhere to the damages sites to regenerate the tissues. Here, comparing normal murine urothelial cells with murine urothelial carcinoma cells (MBT-2), we found that normal cells had less migration ability than carcinoma cells. And in our co-culture system we found that carcinoma cells had propensity migrating toward normal urothelial cells and carcinoma cells had more advantages to adhere than normal cells. To reverse this condition, we used anabolic androgen, dihyrotestosterone (DHT) to treat normal cells and found that DHT treatment increased the migration ability of normal urothelial cells toward carcinoma cells and the adhesion capacity in competition with carcinoma cells. This study provides the base of a novel therapeutic approach by using anabolic hormone-enforced normal urothelial cells to regenerate the damage urothelium and defend against the occupancy of carcinoma cells to thwart cancer development and recurrence.
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW This review addresses significant changes in our understanding of urothelial development and regeneration. Understanding urothelial differentiation will be important in the push to find new methods of bladder reconstruction and augmentation, as well as identification of bladder cancer stem cells. RECENT FINDINGS This review will cover recent findings including the identification of novel progenitor cells in the embryo and adult urothelium, function of the urothelium, and regeneration of the urothelium. Using Cre-lox recombination with cell-type-specific Cre lines, lineage studies from our laboratory have revealed novel urothelial cell types and progenitors that are critical for formation and regeneration of the urothelium. Interestingly, our studies indicate that Keratin-5-expressing basal cells, which have previously been proposed to be urothelial stem cells, are a self-renewing unipotent population, whereas P-cells, a novel urothelial cell type, are progenitors in the embryo, and intermediate cells serve as a progenitor pool in the adult. SUMMARY These findings could have important implications for our understanding of cancer tumorigenesis and could move the fields of regeneration and reconstruction forward.
Collapse
|
19
|
Urine as a source of stem cells. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2014; 129:19-32. [PMID: 23038280 DOI: 10.1007/10_2012_157] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Traditionally, clinicians and researchers have relied on a skin biopsy or blood extraction as relatively accessible supplies for in vitro cell expansion and biological studies. Perhaps surprisingly, limited attention has been given to a totally noninvasive source, urine, which eliminates the discomfort associated with other procedures. This may arise from the perception that urine is merely a body waste. Yet, the analysis of urine is a longstanding fundamental test for diagnostic purposes and nowadays there is growing interest in using urine for detecting biomarkers. In addition, recent work including ours reinforces the idea that urine contains a variety of viable cell types with relevant applications. In this review, we describe those cell types and their potential uses.
Collapse
|
20
|
Generation of bladder urothelium from human pluripotent stem cells under chemically defined serum- and feeder-free system. Int J Mol Sci 2014; 15:7139-57. [PMID: 24776760 PMCID: PMC4057664 DOI: 10.3390/ijms15057139] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 03/25/2014] [Accepted: 04/11/2014] [Indexed: 12/11/2022] Open
Abstract
Human stem cells are promising sources for bladder regeneration. Among several possible sources, pluripotent stem cells are the most fascinating because they can differentiate into any cell type, and proliferate limitlessly in vitro. Here, we developed a protocol for differentiation of human pluripotent stem cells (hPSCs) into bladder urothelial cells (BUCs) under a chemically defined culture system. We first differentiated hPSCs into definitive endoderm (DE), and further specified DE cells into BUCs by treating retinoic acid under a keratinocyte-specific serum free medium. hPSC-derived DE cells showed significantly expressed DE-specific genes, but did not express mesodermal or ectodermal genes. After DE cells were specified into BUCs, they notably expressed urothelium-specific genes such as UPIb, UPII, UPIIIa, P63 and CK7. Immunocytochemistry showed that BUCs expressed UPII, CK8/18 and P63 as well as tight junction molecules, E-CADHERIN and ZO-1. Additionally, hPSCs-derived BUCs exhibited low permeability in a FITC-dextran permeability assay, indicating BUCs possessed the functional units of barrier on their surfaces. However, BUCs did not express the marker genes of other endodermal lineage cells (intestine and liver) as well as mesodermal or ectodermal lineage cells. In summary, we sequentially differentiated hPSCs into DE and BUCs in a serum- and feeder-free condition. Our differentiation protocol will be useful for producing cells for bladder regeneration and studying normal and pathological development of the human bladder urothelium in vitro.
Collapse
|
21
|
Osborn SL, Thangappan R, Luria A, Lee JH, Nolta J, Kurzrock EA. Induction of human embryonic and induced pluripotent stem cells into urothelium. Stem Cells Transl Med 2014; 3:610-9. [PMID: 24657961 DOI: 10.5966/sctm.2013-0131] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In vitro generation of human urothelium from stem cells would be a major advancement in the regenerative medicine field, providing alternate nonurologic and/or nonautologous tissue sources for bladder grafts. Such a model would also help decipher the mechanisms of urothelial differentiation and would facilitate investigation of deviated differentiation of normal progenitors into urothelial cancer stem cells, perhaps elucidating areas of intervention for improved treatments. Thus far, in vitro derivation of urothelium from human embryonic stem cells (hESCs) or human induced pluripotent stem (hiPS) cells has not been reported. The goal of this work was to develop an efficient in vitro protocol for the induction of hESCs into urothelium through an intermediary definitive endoderm step and free of matrices and cell contact. During directed differentiation in a urothelial-specific medium ("Uromedium"), hESCs produced up to 60% urothelium, as determined by uroplakin expression; subsequent propagation selected for 90% urothelium. Alteration of the epithelial and mesenchymal cell signaling contribution through noncell contact coculture or conditioned media did not enhance the production of urothelium. Temporospatial evaluation of transcription factors known to be involved in urothelial specification showed association of IRF1, GET1, and GATA4 with uroplakin expression. Additional hESC and hiPS cell lines could also be induced into urothelium using this in vitro system. These results demonstrate that derivation and propagation of urothelium from hESCs and hiPS cells can be efficiently accomplished in vitro in the absence of matrices, cell contact, or adult cell signaling and that the induction process appears to mimic normal differentiation.
Collapse
Affiliation(s)
- Stephanie L Osborn
- Departments of Urology and Internal Medicine, Davis School of Medicine, and Stem Cell Program, Institute for Regenerative Cures, Davis Medical Center, University of California, Sacramento, California, USA
| | | | | | | | | | | |
Collapse
|
22
|
Moad M, Pal D, Hepburn AC, Williamson SC, Wilson L, Lako M, Armstrong L, Hayward SW, Franco OE, Cates JM, Fordham SE, Przyborski S, Carr-Wilkinson J, Robson CN, Heer R. A novel model of urinary tract differentiation, tissue regeneration, and disease: reprogramming human prostate and bladder cells into induced pluripotent stem cells. Eur Urol 2013; 64:753-61. [PMID: 23582880 PMCID: PMC3819995 DOI: 10.1016/j.eururo.2013.03.054] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 03/25/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Primary culture and animal and cell-line models of prostate and bladder development have limitations in describing human biology, and novel strategies that describe the full spectrum of differentiation from foetal through to ageing tissue are required. Recent advances in biology demonstrate that direct reprogramming of somatic cells into pluripotent embryonic stem cell (ESC)-like cells is possible. These cells, termed induced pluripotent stem cells (iPSCs), could theoretically generate adult prostate and bladder tissue, providing an alternative strategy to study differentiation. OBJECTIVE To generate human iPSCs derived from normal, ageing, human prostate (Pro-iPSC), and urinary tract (UT-iPSC) tissue and to assess their capacity for lineage-directed differentiation. DESIGN, SETTING, AND PARTICIPANTS Prostate and urinary tract stroma were transduced with POU class 5 homeobox 1 (POU5F1; formerly OCT4), SRY (sex determining region Y)-box 2 (SOX2), Kruppel-like factor 4 (gut) (KLF4), and v-myc myelocytomatosis viral oncogene homolog (avian) (MYC, formerly C-MYC) genes to generate iPSCs. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The potential for differentiation into prostate and bladder lineages was compared with classical skin-derived iPSCs. The student t test was used. RESULTS AND LIMITATIONS Successful reprogramming of prostate tissue into Pro-iPSCs and bladder and ureter into UT-iPSCs was demonstrated by characteristic ESC morphology, marker expression, and functional pluripotency in generating all three germ-layer lineages. In contrast to conventional skin-derived iPSCs, Pro-iPSCs showed a vastly increased ability to generate prostate epithelial-specific differentiation, as characterised by androgen receptor and prostate-specific antigen induction. Similarly, UT-iPSCs were shown to be more efficient than skin-derived iPSCs in undergoing bladder differentiation as demonstrated by expression of urothelial-specific markers: uroplakins, claudins, and cytokeratin; and stromal smooth muscle markers: α-smooth-muscle actin, calponin, and desmin. These disparities are likely to represent epigenetic differences between individual iPSC lines and highlight the importance of organ-specific iPSCs for tissue-specific studies. CONCLUSIONS IPSCs provide an exciting new model to characterise mechanisms regulating prostate and bladder differentiation and to develop novel approaches to disease modelling. Regeneration of bladder cells also provides an exceptional opportunity for translational tissue engineering.
Collapse
Affiliation(s)
- Mohammad Moad
- Northern Institute for Cancer Research, Newcastle University, UK
| | - Deepali Pal
- Northern Institute for Cancer Research, Newcastle University, UK
| | | | | | - Laura Wilson
- Northern Institute for Cancer Research, Newcastle University, UK
| | - Majlinda Lako
- Institute of Genetic Medicine, Newcastle University, UK
| | | | - Simon W. Hayward
- Department of Urological Surgery, Vanderbilt University Medical Centre, TN, USA
| | - Omar E. Franco
- Department of Urological Surgery, Vanderbilt University Medical Centre, TN, USA
| | - Justin M. Cates
- Department of Urological Surgery, Vanderbilt University Medical Centre, TN, USA
| | - Sarah E. Fordham
- Northern Institute for Cancer Research, Newcastle University, UK
| | | | | | - Craig N. Robson
- Northern Institute for Cancer Research, Newcastle University, UK
| | - Rakesh Heer
- Northern Institute for Cancer Research, Newcastle University, UK
| |
Collapse
|
23
|
Giuliani A, Moroncini F, Mazzoni S, Belicchi MLC, Villa C, Erratico S, Colombo E, Calcaterra F, Brambilla L, Torrente Y, Albertini G, Della Bella S. Polyglycolic acid-polylactic acid scaffold response to different progenitor cell in vitro cultures: a demonstrative and comparative X-ray synchrotron radiation phase-contrast microtomography study. Tissue Eng Part C Methods 2013; 20:308-16. [PMID: 23879738 DOI: 10.1089/ten.tec.2013.0213] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Spatiotemporal interactions play important roles in tissue development and function, especially in stem cell-seeded bioscaffolds. Cells interact with the surface of bioscaffold polymers and influence material-driven control of cell differentiation. In vitro cultures of different human progenitor cells, that is, endothelial colony-forming cells (ECFCs) from a healthy control and a patient with Kaposi sarcoma (an angioproliferative disease) and human CD133+ muscle-derived stem cells (MSH 133+ cells), were seeded onto polyglycolic acid-polylactic acid scaffolds. Three-dimensional (3D) images were obtained by X-ray phase-contrast microtomography (micro-CT) and processed with the Modified Bronnikov Algorithm. The method enabled high spatial resolution detection of the 3D structural organization of cells on the bioscaffold and evaluation of the way and rate at which cells modified the construct at different time points from seeding. The different cell types displayed significant differences in the proliferation rate. In conclusion, X-ray synchrotron radiation phase-contrast micro-CT analysis proved to be a useful and sensitive tool to investigate the spatiotemporal pattern of progenitor cell organization on a bioscaffold.
Collapse
Affiliation(s)
- Alessandra Giuliani
- 1 Dipartimento di Scienze Cliniche Specialistiche e Odontostomatologiche, Università Politecnica delle Marche , Ancona, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Age-related alterations in regeneration of the urinary bladder after subtotal cystectomy. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1585-1595. [PMID: 24012523 DOI: 10.1016/j.ajpath.2013.07.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 07/02/2013] [Accepted: 07/16/2013] [Indexed: 01/21/2023]
Abstract
Prior work documented that surgical removal of approximately 70% of the bladder (subtotal cystectomy) in 12-week-old female rats induced complete functional regeneration of the bladder within 8 weeks. To determine whether animal age affects bladder regeneration, female F344 rats aged 12 weeks (young) and 12 months (old) underwent subtotal cystectomy, and then were evaluated from 1 to 26 weeks after subtotal cystectomy. At 26 weeks after subtotal cystectomy, bladder capacity in young animals was indistinguishable from that in age-matched controls, but bladder capacity in old animals was only approximately 56% of that in age-matched controls. There was no detectable difference in residual volume among treatment groups, but the diminished regeneration in old animals was associated with a corresponding increase in the ratio of residual volume to micturition volume. The majority of old animals exhibited evidence of chronic kidney damage after subtotal cystectomy. Maximal contraction of bladder strips to electrical field stimulation, as well as activation with carbachol, phenylephrine, and KCl, were lower in old than in young animals at 26 weeks after subtotal cystectomy. Immunostaining with proliferating cell nuclear antigen and Von Willebrand factor revealed delayed and/or diminished proliferative and angiogenic responses, respectively, in old animals. These results confirm prior work and suggest that multiple mechanisms may contribute to an age-related decline in the regenerative capacity of the bladder.
Collapse
|
25
|
Islam SS, Mokhtari RB, Kumar S, Maalouf J, Arab S, Yeger H, Farhat WA. Spatio-temporal distribution of Smads and role of Smads/TGF-β/BMP-4 in the regulation of mouse bladder organogenesis. PLoS One 2013; 8:e61340. [PMID: 23620745 PMCID: PMC3631207 DOI: 10.1371/journal.pone.0061340] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 03/07/2013] [Indexed: 11/24/2022] Open
Abstract
Although Shh, TGF-β and BMP-4 regulate radial patterning of the bladder mesenchyme and smooth muscle differentiation, it is not known what transcription factors, local environmental cues or signaling cascades mediate bladder smooth muscle differentiation. We investigated the expression patterns of signaling mediated by Smad2 and Smad3 in the mouse embryonic bladder from E12.5 to E16.5 by using qRT-PCR, in situ hybridization and antibodies specifically recognizing individual Smad proteins. The role of Smad2 and Smad3 during smooth muscle formation was examined by disrupting the Smad2/3 signaling pathway using TβR1 inhibitor SB-431542 in organ culture system. qRT-PCR results showed that R-Smads, Co-Smad and I-Smads were all expressed during bladder development. RNA ISH for BMP-4 and immunostaining of TGF-β1 showed that BMP-4 and TGF-β1 were expressed in the transitional epithelium, lamina propia and muscularis mucosa. Smad1, Smad5 and Smad8 were first expressed in the bladder epithelium and continued to be expressed in the transitional epithelium, muscularis mesenchyme and lamina propia as the bladder developed. Smad2, Smad3 and Smad4 were first detected in the bladder epithelium and subsequently were expressed in the muscularis mesenchyme and lamina propia. Smad6 and Smad7 showed overlapping expression with R-Smads, which are critical for bladder development. In bladder explants (E12.5 to E16.5) culture, Smad2 and Smad3 were found localized within the nuclei, suggesting critical transcriptional regulatory effects during bladder development. E12.5 to E16.5 bladders were cultured with and without TβR1 inhibitor SB-431542 and assessed by qRT-PCR and immunofluorescence. After three days in culture in SB-431542, α-SMA, Smad2 and Smad3 expressions were significantly decreased compared with controls, however, with no significant changes in the expression of smooth muscle myosin heavy chain (SM-Myh. Based on the Smad expression patterns, we suggest that individual or combinations of Smads may be necessary during mouse bladder organogenesis and may be critical mediators for bladder smooth muscle differentiation.
Collapse
Affiliation(s)
- Syed S. Islam
- Developmental and Stem Cell Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Reza Bayat Mokhtari
- Department of Pediatric Laboratory Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Sushil Kumar
- Physiology and Experimental Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | | | - Sara Arab
- University of Toronto, Toronto, ON, Canada
| | - Herman Yeger
- Department of Pediatric Laboratory Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Walid A. Farhat
- Developmental and Stem Cell Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| |
Collapse
|
26
|
Zhang M, Peng Y, Zhou Z, Zhou J, Wang Z, Lu M. Differentiation of human adipose-derived stem cells co-cultured with urothelium cell line toward a urothelium-like phenotype in a nude murine model. Urology 2013; 81:465.e15-22. [PMID: 23374843 DOI: 10.1016/j.urology.2012.10.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Revised: 10/01/2012] [Accepted: 10/17/2012] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To investigated the urothelium differentiation potential of adipose-derived stem cells (ASCs) that were coimplanted with the immortalized human bladder urothelium cell line (SV-HUC-1) into the subcutaneous tissue of athymic mice. MATERIALS AND METHODS The ASCs were isolated from the human adipose tissue of patients undergoing liposuction procedures and were expanded in vitro. After labeling with CM-DiI, the ASCs were mixed with SV-HUC-1 and implanted into the subcutaneous tissue of athymic mice for 2 and 4 weeks. The urothelium-specific markers uroplakin-Ia and uroplakin-II were detected by immunofluorescence. The transformation rate of ASCs into the urothelium phenotype was evaluated at each measurement point. RESULTS We found that 25.87% ± 1.38% of ASCs expressed the urothelium-specific marker uroplakin-Ia and 23.60% ± 2.57% of ASCs expressed uroplakin-II 2 weeks after coimplantation with SV-HUC-1 in vivo. After 4 weeks, 70.07% ± 3.84% of ASCs expressed uroplakin-Ia and 65.56% ± 2.94% expressed uroplakin-II. However, no obvious organizational multilayered urothelium structure, such as that of the native bladder mucosa, was found in the subcutaneous tissues of the athymic mice. CONCLUSION The results of our study have demonstrated that ASCs could be differentiated toward the urothelium-like phenotype when they were coimplanted in direct contact with cells of a mature urothelium cell line, and the proportion of differentiated cells increased from 2 to 4 weeks. The differentiation potential of ASCs toward the urothelial cell type suggests that ASCs might have potential to be used in urinary tract repair with a tissue engineering approach in the future.
Collapse
Affiliation(s)
- Ming Zhang
- Department of Urology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | | | |
Collapse
|
27
|
Zhidkova OV, Petrov NS, Popov BV. Preparation and characteristics of growth and marker properties of urinary bladder mesenchymal stem cells. J EVOL BIOCHEM PHYS+ 2013. [DOI: 10.1134/s0022093013010131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
28
|
Zhang Y, Atala A. Urothelial cell culture: stratified urothelial sheet and three-dimensional growth of urothelial structure. Methods Mol Biol 2013; 945:383-99. [PMID: 23097119 DOI: 10.1007/978-1-62703-125-7_23] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Urothelial cells line the urinary tract, including the renal pelvis, ureters, bladder, superior urethra, and the central ducts of the prostate. They are highly specialized epithelial cell types possessing unique features, imparting important functional roles in the urinary system. They act as a permeability barrier and protect underlying muscle tissues from the caustic effects of urine while also expanding with bladder filling to adjust urine pressures. The multilayered urothelium is typically structured with differentiated, mature surface cells and less mature basal cells. The basal cell layer contains tissue-specific stem cells able to self-renew for the lifetime of the mammal and also produces a pool of maturing cells for tissue homeostasis. Maintaining regenerative basal cells in a culture facilitates urothelial cell growth in vitro. Additionally, epithelial-mesenchymal communication, epithelial-matrix interactions, and cytokines/growth factors are required to maintain the normal structure and function of mature urothelial cells in vitro and to induce stem cell differentiation into urothelial cells. These cultures are useful to study the biology and physiology of the urinary tract, particularly for the development of cell-based tissue engineering strategies in urology. This chapter describes methods for the isolation of urothelial cells and their maintenance in monolayer culture, and methods for the production of multilayer urothelial cell sheets and three-dimensional cocultures of urothelial and mesenchymal cells.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Urology, Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | | |
Collapse
|
29
|
Abstract
This chapter reviews the use of urothelial cells as a means to enhance tissue regeneration and wound healing in urinary tract system. It addresses the properties of urothelial cells, including their role as a permeability barrier to protect underlying muscle tissue from the caustic effects of urine and as one of the main cell types, along with smooth muscle cells, that are used in urethral or bladder tissue engineering today. This description includes a general overview of various isolation techniques and culture methods that have been developed to improve urinary tract reconstruction in vivo and aid the characterization of growth factor expression in vitro. The chapter then describes various applications using urothelial cells, including production of multilayer urothelial sheets, tissue engineered bladder mucosa, tissue engineered urethra, and tissue engineered bladder. It also outlines the advantages of sandwich and layered coculture of these cells and the effects of epithelial-stromal cell interactions during tissue regeneration or wound healing processes in the urinary tract.
Collapse
|
30
|
Zhou L, Yang B, Sun C, Qiu X, Sun Z, Chen Y, Zhang Y, Dai Y. Coadministration of platelet-derived growth factor-BB and vascular endothelial growth factor with bladder acellular matrix enhances smooth muscle regeneration and vascularization for bladder augmentation in a rabbit model. Tissue Eng Part A 2012; 19:264-76. [PMID: 22894544 DOI: 10.1089/ten.tea.2011.0609] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Tissue-engineering techniques have brought a great hope for bladder repair and reconstruction. The crucial requirements of a tissue-engineered bladder are bladder smooth muscle regeneration and vascularization. In this study, partial rabbit bladder (4×5 cm) was removed and replaced with a porcine bladder acellular matrix (BAM) that was equal in size. BAM was incorporated with platelet-derived growth factor-BB (PDGF-BB) and vascular endothelial growth factor (VEGF) in the experimental group while with no bioactive factors in the control group. The bladder tissue strip contractility in the experimental rabbits was better than that in the control ones postoperation. Histological evaluation revealed that smooth muscle regeneration and vascularization in the experimental group were significantly improved compared with those in the control group (p<0.05), while multilayered urothelium was formed in both groups. Muscle strip contractility of neobladder in the experimental group exhibited significantly better than that in the control (p<0.05) assessed with electrical field stimulation and carbachol interference. The activity of matrix metalloproteinase-2 (MMP-2) and MMP-9 in the native bladder tissue around tissue-engineered neobladder in the experimental group was significantly higher than that in the control (p<0.05). This work suggests that smooth muscle regeneration and vascularization in tissue-engineered neobladder and recovery of bladder function could be enhanced by PDGF-BB and VEGF incorporated within BAM, which promoted the upregulation of the activity of MMP-2 and MMP-9 of native bladder tissue around the tissue-engineered neobladder.
Collapse
Affiliation(s)
- Liuhua Zhou
- Department of Urology, Nanjing University School of Medicine, Affiliated Drum Tower Hospital, Nanjing, China
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Vaegler M, Lenis AT, Daum L, Amend B, Stenzl A, Toomey P, Renninger M, Damaser MS, Sievert KD. Stem cell therapy for voiding and erectile dysfunction. Nat Rev Urol 2012; 9:435-47. [PMID: 22710667 PMCID: PMC3769422 DOI: 10.1038/nrurol.2012.111] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Voiding dysfunction comprises a variety of disorders, including stress urinary incontinence and overactive bladder, and affects millions of men and women worldwide. Erectile dysfunction (ED) also decreases quality of life for millions of men, as well as for their partners. Advanced age and diabetes are common comorbidities that can exacerbate and negatively impact upon the development of these disorders. Therapies that target the pathophysiology of these conditions to halt progression are not currently available. However, stem cell therapy could fill this therapeutic void. Stem cells can reduce inflammation, prevent fibrosis, promote angiogenesis, recruit endogenous progenitor cells, and differentiate to replace damaged cells. Adult multipotent stem cell therapy, in particular, has shown promise in case reports and preclinical animal studies. Stem cells also have a role in urological tissue engineering for ex vivo construction of bladder wall and urethral tissue (using a patient's own cells) prior to transplantation. More recent studies have focused on bioactive factor secretion and homing of stem cells. In the future, clinicians are likely to utilize allogeneic stem cell sources, intravenous systemic delivery, and ex vivo cell enhancement to treat voiding dysfunction and ED.
Collapse
Affiliation(s)
- Martin Vaegler
- Department of Urology, University of Tuebingen, Hoppe-Seyler-Strasse 3, D72076 Tuebingen, Germany
| | - Andrew T Lenis
- The Cleveland Clinic, Case Western Reserve University School of Medicine, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Lisa Daum
- Department of Urology, University of Tuebingen, Hoppe-Seyler-Strasse 3, D72076 Tuebingen, Germany
| | - Bastian Amend
- Department of Urology, University of Tuebingen, Hoppe-Seyler-Strasse 3, D72076 Tuebingen, Germany
| | - Arnulf Stenzl
- Department of Urology, University of Tuebingen, Hoppe-Seyler-Strasse 3, D72076 Tuebingen, Germany
| | - Patricia Toomey
- Department of Urology, University of Tuebingen, Hoppe-Seyler-Strasse 3, D72076 Tuebingen, Germany
| | - Markus Renninger
- Department of Urology, University of Tuebingen, Hoppe-Seyler-Strasse 3, D72076 Tuebingen, Germany
| | - Margot S Damaser
- The Cleveland Clinic, Case Western Reserve University School of Medicine, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Karl-Dietrich Sievert
- Department of Urology, University of Tuebingen, Hoppe-Seyler-Strasse 3, D72076 Tuebingen, Germany
| |
Collapse
|
32
|
Liu AY, Vêncio RZN, Page LS, Ho ME, Loprieno MA, True LD. Bladder expression of CD cell surface antigens and cell-type-specific transcriptomes. Cell Tissue Res 2012; 348:589-600. [PMID: 22427119 DOI: 10.1007/s00441-012-1383-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 02/23/2012] [Indexed: 12/13/2022]
Abstract
Many cell types have no known functional attributes. In the bladder and prostate, basal epithelial and stromal cells appear similar in cytomorphology and share several cell surface markers. Their total gene expression (transcriptome) should provide a clear measure of the extent to which they are alike functionally. Since urologic stromal cells are known to mediate organ-specific tissue formation, these cells in cancers might exhibit aberrant gene expression affecting their function. For transcriptomes, cluster designation (CD) antigens have been identified for cell sorting. The sorted cell populations can be analyzed by DNA microarrays. Various bladder cell types have unique complements of CD molecules. CD9(+) urothelial, CD104(+) basal and CD13(+) stromal cells of the lamina propria were therefore analyzed, as were CD9(+) cancer and CD13(+) cancer-associated stromal cells. The transcriptome datasets were compared by principal components analysis for relatedness between cell types; those with similarity in gene expression indicated similar function. Although bladder and prostate basal cells shared CD markers such as CD104, CD44 and CD49f, they differed in overall gene expression. Basal cells also lacked stem cell gene expression. The bladder luminal and stromal transcriptomes were distinct from their prostate counterparts. In bladder cancer, not only the urothelial but also the stromal cells showed gene expression alteration. The cancer process in both might thus involve defective stromal signaling. These cell-type transcriptomes provide a means to monitor in vitro models in which various CD-isolated cell types can be combined to study bladder differentiation and bladder tumor development based on cell-cell interaction.
Collapse
Affiliation(s)
- Alvin Y Liu
- Department of Urology and Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Selenoproteins in bladder cancer. Clin Chim Acta 2012; 413:847-54. [PMID: 22349600 DOI: 10.1016/j.cca.2012.01.041] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Revised: 01/19/2012] [Accepted: 01/31/2012] [Indexed: 12/13/2022]
Abstract
Selenoproteins with genetically encoded selenium (Se) are very important in response to oxidative stress, redox balance and regulation of various metabolic and developmental processes. Although increased circulating Se has been associated with 33% risk reduction of bladder cancer, there are little data on selenoprotein expression at the protein and genetic level from both human and animal studies. Data from the Mammalian Gene Collection (MGC) Project clearly showed that highest mRNA expression in human urinary epithelium for TRXR1 (thioredoxin reductase 1), GPX1 (glutathione peroxidase 1), SEP15 (15 kDa selenoprotein), SELT (selenoprotein T) and SEPW1 (selenoprotein W1). Although bladder tumor has been characterized by increased Se, GPX and TRXR activity, circulating Se and GPX was interestingly decreased in these cancer patients. As such, selenoprotein expression in urinary epithelium may be involved in bladder cancer (development, progression and recurrence) and may play a significant role in chemotherapeutic intervention. Despite these findings, the role of selenoproteins in bladder cancer has rarely been investigated and the significance of selenoproteins in normal and malignant uroepithelium remains poorly understood.
Collapse
|
34
|
Abstract
Organotypic culture is an invaluable technique that allows researchers with the tool to analyze a tissue development in an isolated and well-defined environment. This technique also permits one to study the roles of different signaling systems/signaling molecules and to take advantage of the modern real-time imaging techniques, including confocal microscopy. With great success, our lab has used organotypic culture of the urogenital tract (UGT) to study growth and extension of the mesonephric (Wolffian) duct and its cloaca connection, ureter maturation, and bladder urothelium development (Batourina et al. Nat Genet 32:109, 2002; Batourina et al. Nat Genet 37:1082, 2005; Mendelsohn Organogenesis 5:306, 2009).
Collapse
|
35
|
Azman J, Starcevic Klasan G, Ivanac D, Picard A, Jurisic-Erzen D, Nikolic M, Malnar D, Arbanas J, Jerkovic R. Reg IV protein and mRNA expression in different rat organs. Acta Histochem 2011; 113:793-7. [PMID: 21168191 DOI: 10.1016/j.acthis.2010.11.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Revised: 11/24/2010] [Accepted: 11/25/2010] [Indexed: 12/20/2022]
Abstract
The Reg IV gene has been documented in the human colon, small intestine, stomach and pancreas. Expression of the Reg IV in different cell types has been associated with regeneration, cell growth and cell survival, cell adhesion and resistance to apoptosis. Since the distribution of the Reg IV protein in normal rat tissues is unknown, the aim of this study was to reveal the expression of the Reg IV protein in structurally and functionally different rat organs. The expression of Reg IV gene was analyzed by Western blot and reverse transcription-polymerase chain reaction. Immunohistochemistry was used to localize Reg IV protein. Reg IV protein was expressed in pancreas, stomach, small intestine, colon, brain, spleen, kidney and urinary bladder in two-month-old male Wistar rats. In addition, the expression of Reg IV mRNA by reverse transcription-polymerase chain reaction was confirmed. Our study provides detailed information about the expression and localization of Reg IV protein in different rat organs. These findings provide an evidence of Reg IV expression in different rat organs, which may help elucidate a potential role in growth and proliferation of different cells like other members of the Reg family genes which act as growth factors in the different organs.
Collapse
Affiliation(s)
- Josip Azman
- Department of Anesthesiology and Intensive Care, University Hospital Rijeka, Kresimirova 42, Rijeka, Croatia
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Yang B, Zhang Y, Zhou L, Sun Z, Zheng J, Chen Y, Dai Y. Development of a porcine bladder acellular matrix with well-preserved extracellular bioactive factors for tissue engineering. Tissue Eng Part C Methods 2011; 16:1201-11. [PMID: 20170425 DOI: 10.1089/ten.tec.2009.0311] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In this study, we compared four decellularization protocols and finally developed an optimized one through which a porcine bladder acellular matrix (BAM) with well-preserved extracellular bioactive factors had been prepared. In this protocol, the intact bladder was treated with trypsin/ethylenediaminetetraacetic acid to remove the urothelium, then with hypotonic buffer and Triton X-100 in hypertonic buffer to remove the membranous and cytoplasmic materials, and finally with nuclease to degrade the cellular nuclear components. Bladder distention and mechanical agitation were simultaneously used to facilitate cell removal. Meanwhile, several preservative techniques, including limitation of wash time, supplement with inhibitors of proteinase, control of the pH value and temperature of the wash buffer, ethylene oxide sterilization, and lyophilization of the scaffold for storage, were used to protect the extracellular bioactive factors. This decellularization protocol had completely removed the cellular materials and well preserved the extracellular collagen, sulfated glycosaminoglycan (GAG), and bioactive factors. The preserved bioactive factors had a great potential of promoting the proliferation and migration of both human bladder smooth muscle cell and human umbilical vein endothelial cell. It was also found that the amount of two representative bioactive factors, platelet-derived growth factor BB and vascular endothelial growth factor, was positively correlated with the sulfated GAG content in the porcine BAM, implying that the amount of sulfated GAG might be a determinant for preservation of bioactive factors in the decellularized tissues. In conclusion, the porcine BAM with well-preserved extracellular bioactive factors might be a favorable scaffold for tissue engineering applications.
Collapse
Affiliation(s)
- Bin Yang
- Department of Urology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, PR China
| | | | | | | | | | | | | |
Collapse
|
37
|
Mauney JR, Ramachandran A, Yu RN, Daley GQ, Adam RM, Estrada CR. All-trans retinoic acid directs urothelial specification of murine embryonic stem cells via GATA4/6 signaling mechanisms. PLoS One 2010; 5:e11513. [PMID: 20644631 PMCID: PMC2903484 DOI: 10.1371/journal.pone.0011513] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Accepted: 06/02/2010] [Indexed: 01/05/2023] Open
Abstract
The urinary bladder and associated tract are lined by the urothelium, a transitional epithelium that acts as a specialized permeability barrier that protects the underlying tissue from urine via expression of a highly specific group of proteins known as the uroplakins (UP). To date, our understanding of the developmental processes responsible for urothelial differentiation has been hampered due to the lack of suitable models. In this study, we describe a novel in vitro cell culture system for derivation of urothelial cells from murine embryonic stem cells (ESCs) following cultivation on collagen matrices in the presence all trans retinoic acid (RA). Upon stimulation with micromolar concentrations of RA, ESCs significantly downregulated the pluripotency factor OCT-4 but markedly upregulated UP1A, UP1B, UP2, UP3A, and UP3B mRNA levels in comparison to naïve ESCs and spontaneously differentiating controls. Pan-UP protein expression was associated with both p63- and cytokeratin 20-positive cells in discrete aggregating populations of ESCs following 9 and 14 days of RA stimulation. Analysis of endodermal transcription factors such as GATA4 and GATA6 revealed significant upregulation and nuclear enrichment in RA-treated UP2-GFP+ populations. GATA4-/- and GATA6-/- transgenic ESC lines revealed substantial attenuation of RA-mediated UP expression in comparison to wild type controls. In addition, EMSA analysis revealed that RA treatment induced formation of transcriptional complexes containing GATA4/6 on both UP1B and UP2 promoter fragments containing putative GATA factor binding sites. Collectively, these data suggest that RA mediates ESC specification toward a urothelial lineage via GATA4/6-dependent processes.
Collapse
Affiliation(s)
- Joshua R. Mauney
- Urological Diseases Research Center, Children's Hospital Boston, Boston, Massachusetts, United States of America
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Aruna Ramachandran
- Urological Diseases Research Center, Children's Hospital Boston, Boston, Massachusetts, United States of America
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Richard N. Yu
- Urological Diseases Research Center, Children's Hospital Boston, Boston, Massachusetts, United States of America
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - George Q. Daley
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
- Harvard Stem Cell Institute, Boston, Massachusetts, United States of America
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Children's Hospital Boston, Boston, Massachusetts, United States of America
- Dana Farber Cancer Institute, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
- Division of Hematology, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Manton Center for Orphan Disease Research, Boston, Massachusetts, United States of America
| | - Rosalyn M. Adam
- Urological Diseases Research Center, Children's Hospital Boston, Boston, Massachusetts, United States of America
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Carlos R. Estrada
- Urological Diseases Research Center, Children's Hospital Boston, Boston, Massachusetts, United States of America
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
38
|
Lu M, Zhou G, Liu W, Wang Z, Zhu Y, Yu B, Zhang W, Cao Y. Remodeling of Buccal Mucosa by Bladder Microenvironment. Urology 2010; 75:1514.e7-14. [DOI: 10.1016/j.urology.2009.12.060] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Revised: 12/01/2009] [Accepted: 12/22/2009] [Indexed: 10/19/2022]
|
39
|
Ashley RA, Palmer BW, Schultz AD, Woodson BW, Roth CC, Routh JC, Fung KM, Frimberger D, Lin HK, Kropp BP. Leukocyte Inflammatory Response in a Rat Urinary Bladder Regeneration Model Using Porcine Small Intestinal Submucosa Scaffold. Tissue Eng Part A 2009; 15:3241-6. [DOI: 10.1089/ten.tea.2008.0699] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Richard A. Ashley
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Blake W. Palmer
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Andrew D. Schultz
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Benjamin W. Woodson
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Christopher C. Roth
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Jonathan C. Routh
- Harvard School of Public Health, Harvard Medical School, Boston, Massachusetts
| | - Kar-Ming Fung
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- U.S. Department of Veterans Affairs Medical Center, Oklahoma City, Oklahoma
| | - Dominic Frimberger
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Hsueh-Kung Lin
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- U.S. Department of Veterans Affairs Medical Center, Oklahoma City, Oklahoma
| | - Bradley P. Kropp
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
40
|
Reyes L, Alvarez S, Allam A, Reinhard M, Brown MB. Complicated urinary tract infection is associated with uroepithelial expression of proinflammatory protein S100A8. Infect Immun 2009; 77:4265-74. [PMID: 19667050 PMCID: PMC2747944 DOI: 10.1128/iai.00458-09] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Revised: 06/05/2009] [Accepted: 07/30/2009] [Indexed: 11/20/2022] Open
Abstract
F344 rats chronically infected with Ureaplasma parvum develop two distinct profiles: asymptomatic urinary tract infection (UTI) and UTI complicated by struvite urolithiasis. To identify factors that affect disease outcome, we characterized the temporal host immune response during infection by histopathologic analysis and in situ localization of U. parvum. We also used differential quantitative proteomics to identify distinguishing host cellular responses associated with complicated UTI. In animals in which microbial colonization was limited to the mucosal surface, inflammation was indistinguishable from that which occurred in sham-inoculated controls, and the inflammation resolved by 72 h postinoculation (p.i.) in both groups. However, inflammation persisted in animals with microbial colonization that extended into the deeper layers of the submucosa. Proteome profiling showed that bladder tissues from animals with complicated UTIs had significant increases (P < 0.01) in proteins involved in apoptosis, oxidative stress, and inflammation. Animals with complicated UTIs (2 weeks p.i.) had the highest concentrations of the proinflammatory protein S100A8 (P
Collapse
Affiliation(s)
- Leticia Reyes
- University of Florida, College of Veterinary Medicine, Department of Infectious Diseases & Pathology, P.O. Box 110880, Gainesville, FL 32611-0880, USA.
| | | | | | | | | |
Collapse
|
41
|
Liu B, Cunha GR, Baskin LS. Differential expression of microRNAs in mouse embryonic bladder. Biochem Biophys Res Commun 2009; 385:528-33. [DOI: 10.1016/j.bbrc.2009.05.088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2009] [Accepted: 05/20/2009] [Indexed: 10/20/2022]
|
42
|
Jack GS, Zhang R, Lee M, Xu Y, Wu BM, Rodríguez LV. Urinary bladder smooth muscle engineered from adipose stem cells and a three dimensional synthetic composite. Biomaterials 2009; 30:3259-70. [PMID: 19345408 DOI: 10.1016/j.biomaterials.2009.02.035] [Citation(s) in RCA: 155] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2008] [Accepted: 02/24/2009] [Indexed: 12/15/2022]
Abstract
Human adipose stem cells were cultured in smooth muscle inductive media and seeded into synthetic bladder composites to tissue engineer bladder smooth muscle. 85:15 Poly-lactic-glycolic acid bladder dome composites were cast using an electropulled microfiber luminal surface combined with an outer porous sponge. Cell-seeded bladders expressed smooth muscle actin, myosin heavy chain, calponinin, and caldesmon via RT-PCR and immunoflourescence. Nude rats (n=45) underwent removal of half their bladder and repair using: (i) augmentation with the adipose stem cell-seeded composites, (ii) augmentation with a matched acellular composite, or (iii) suture closure. Animals were followed for 12 weeks post-implantation and bladders were explanted serially. Results showed that bladder capacity and compliance were maintained in the cell-seeded group throughout the 12 weeks, but deteriorated in the acellular scaffold group sequentially with time. Control animals repaired with sutures regained their baseline bladder capacities by week 12, demonstrating a long-term limitation of this model. Histological analysis of explanted materials demonstrated viable adipose stem cells and increasing smooth muscle mass in the cell-seeded scaffolds with time. Tissue bath stimulation demonstrated smooth muscle contraction of the seeded implants but not the acellular implants after 12 weeks in vivo. Our study demonstrates the feasibility and short term physical properties of bladder tissue engineered from adipose stem cells.
Collapse
Affiliation(s)
- Gregory S Jack
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, United States
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
Urothelium covers the inner surfaces of the renal pelvis, ureter, bladder, and prostatic urethra. Although morphologically similar, the urothelia in these anatomic locations differ in their embryonic origin and lineages of cellular differentiation, as reflected in their different uroplakin content, expandability during micturition, and susceptibility to chemical carcinogens. Previously thought to be an inert tissue forming a passive barrier between the urine and blood, urothelia have recently been shown to have a secretory activity that actively modifies urine composition. Urothelial cells express a number of ion channels, receptors, and ligands, enabling them to receive and send signals and communicate with adjoining cells and their broader environment. The urothelial surface bears specific receptors that not only allow uropathogenic E. coli to attach to and invade the bladder mucosa, but also provide a route by which the bacteria ascend through the ureters to the kidney to cause pyelonephritis. Genetic ablation of one or more uroplakin genes in mice causes severe retrograde vesicoureteral reflux, hydronephrosis, and renal failure, conditions that mirror certain human congenital diseases. Clearly, abnormalities of the lower urinary tract can impact the upper tract, and vice versa, through the urothelial connection. In this review, we highlight recent advances in the field of urothelial biology by focusing on the uroplakins, a group of urothelium-specific and differentiation-dependent integral membrane proteins. We discuss these proteins' biochemistry, structure, assembly, intracellular trafficking, and their emerging roles in urothelial biology, function, and pathological processes. We also call attention to important areas where greater investigative efforts are warranted.
Collapse
|
44
|
Goo YA, Liu AY, Ryu S, Shaffer SA, Malmström L, Page L, Nguyen LT, Doneanu CE, Goodlett DR. Identification of secreted glycoproteins of human prostate and bladder stromal cells by comparative quantitative proteomics. Prostate 2009; 69:49-61. [PMID: 18792917 PMCID: PMC4281891 DOI: 10.1002/pros.20853] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Functional development of the prostate is governed by stromal mesenchyme induction and epithelial response. Stromal/epithelial signaling can be mediated through direct cell-cell contact and diffusible factors and their cell surface receptors. These inducers are likely secreted or membrane-associated extracellular proteins. Given the importance of intercellular communication, it is possible that diseases like cancer could arise from a loss of this communication. One approach to gain a molecular understanding of stromal cells is to identify, as a first step, secreted stromal signaling factors. We proposed to do this by comparative analysis between bladder and prostate. METHODS Secreted proteins were identified from cultured normal prostate and bladder stromal mesenchyme cells by glycopeptide-capture method followed by mass spectrometry. Differences in protein abundance between prostate and bladder were quantified from calculated peptide ion current area (PICA) followed by Western validation. Functional and pathway analyses of the proteins were carried out by Gene Ontology (GO) and Teranode software. RESULTS This analysis produced a list of 116 prostate and 84 bladder secreted glycoproteins with ProteinProphet probability scores > or =0.9. Stromal proteins upregulated in the prostate include cathepsin L, follistatin-related protein, neuroendocrine convertase, tumor necrosis factor receptor, and others that are known to be involved in signal transduction, extracellular matrix interaction, differentiation and transport. CONCLUSIONS We have identified a number of potential proteins for stromal signaling and bladder or prostate differentiation program. The prostate stromal/epithelial signaling may be accomplished through activation of the ECM-receptor interaction, complement and coagulation cascades, focal adhesion and cell adhesion pathways.
Collapse
Affiliation(s)
- Young Ah Goo
- Urology (Box 356510), University of Washington, Seattle, WA 98195
- Medicinal Chemistry (Box 357610), University of Washington, Seattle, WA 98195
- Institute for Systems Biology, 1441 N. 34 St. Seattle, WA, 98103
- Corresponding Authors: University of Washington, Medicinal Chemistry, Box 357610, Seattle, WA 98195-7610, USA, Tel: 206-616-4586, Fax: 206-685-3252, ,
| | - Alvin Y. Liu
- Urology (Box 356510), University of Washington, Seattle, WA 98195
- Institute for Systems Biology, 1441 N. 34 St. Seattle, WA, 98103
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195
| | - Soyoung Ryu
- Medicinal Chemistry (Box 357610), University of Washington, Seattle, WA 98195
| | - Scott A. Shaffer
- Medicinal Chemistry (Box 357610), University of Washington, Seattle, WA 98195
| | - Lars Malmström
- Medicinal Chemistry (Box 357610), University of Washington, Seattle, WA 98195
| | - Laura Page
- Urology (Box 356510), University of Washington, Seattle, WA 98195
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195
| | - Liem T. Nguyen
- Medicinal Chemistry (Box 357610), University of Washington, Seattle, WA 98195
| | | | - David R. Goodlett
- Medicinal Chemistry (Box 357610), University of Washington, Seattle, WA 98195
- Institute for Systems Biology, 1441 N. 34 St. Seattle, WA, 98103
- Corresponding Authors: University of Washington, Medicinal Chemistry, Box 357610, Seattle, WA 98195-7610, USA, Tel: 206-616-4586, Fax: 206-685-3252, ,
| |
Collapse
|
45
|
Funk GA, Gosert R, Comoli P, Ginevri F, Hirsch HH. Polyomavirus BK replication dynamics in vivo and in silico to predict cytopathology and viral clearance in kidney transplants. Am J Transplant 2008; 8:2368-77. [PMID: 18925904 DOI: 10.1111/j.1600-6143.2008.02402.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Fast BK virus (BKV) replication in renal tubular epithelial cells drives polyomavirus-BK-associated nephropathy (PVAN) to premature kidney transplant (KT) failure. BKV also replicates in urothelial cells, but remains asymptomatic in two-thirds of affected KT patients. Comparing 518 day-matched plasma-urine samples from 223 KT patients, BKV loads were approximately 3000-fold higher in urine than in plasma (p < 0.000001). Molecular and quantitative parameters indicated that >95% of urine BKV loads resulted from urothelial replication and <5% from tubular epithelial replication. Fast BKV replication dynamics in plasma and urine with half-lives of <12 h accounted for daily urothelial and tubular epithelial cell loss of 4 x 10(7) and 6 x 10(7), respectively. BKV dynamics in both sites were only partly linked, with full and partial discordance in 36% and 32%, respectively. Viral expansion was best explained by models where BKV replication started in the kidney followed by urothelial amplification and tubular epithelial cell cross-feeding reaching a dynamic equilibrium after approximately 10 weeks. Curtailing intrarenal replication by 50% was ineffective and >80% was required for clearing viremia within 7 weeks, but viruria persisted for >14 weeks. Reductions >90% cleared viremia and viruria by 3 and 10 weeks, respectively. The model was clinically validated in prospectively monitored KT patients supporting >80% curtailing for optimal interventions.
Collapse
Affiliation(s)
- G A Funk
- Transplantation Virology, Institute for Medical Microbiology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | | | | | | |
Collapse
|
46
|
Thomas JC, Oottamasathien S, Makari JH, Honea L, Sharif-Afshar AR, Wang Y, Adams C, Wills ML, Bhowmick NA, Adams MC, Brock JW, Hayward SW, Matusik RJ, Pope JC. Temporal-Spatial Protein Expression in Bladder Tissue Derived From Embryonic Stem Cells. J Urol 2008; 180:1784-9. [DOI: 10.1016/j.juro.2008.03.098] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2007] [Indexed: 11/26/2022]
Affiliation(s)
- John C. Thomas
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Pediatric Urology, Monroe Carell Jr. Vanderbilt Children's Hospital, Nashville, Tennessee
| | - Siam Oottamasathien
- Division of Pediatric Urology, Primary Children's Medical Center, Salt Lake City, Utah
| | - John H. Makari
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Pediatric Urology, Monroe Carell Jr. Vanderbilt Children's Hospital, Nashville, Tennessee
| | - Lindsay Honea
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ali-Reza Sharif-Afshar
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yongqing Wang
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Cyrus Adams
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Marcia L. Wills
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Neil A. Bhowmick
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Pediatric Urology, Monroe Carell Jr. Vanderbilt Children's Hospital, Nashville, Tennessee
| | - Mark C. Adams
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Pediatric Urology, Monroe Carell Jr. Vanderbilt Children's Hospital, Nashville, Tennessee
| | - John W. Brock
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Pediatric Urology, Monroe Carell Jr. Vanderbilt Children's Hospital, Nashville, Tennessee
| | - Simon W. Hayward
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Robert J. Matusik
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - John C. Pope
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Pediatric Urology, Monroe Carell Jr. Vanderbilt Children's Hospital, Nashville, Tennessee
| |
Collapse
|
47
|
Anumanthan G, Makari JH, Honea L, Thomas JC, Wills ML, Bhowmick NA, Adams MC, Hayward SW, Matusik RJ, Brock JW, Pope JC. Directed differentiation of bone marrow derived mesenchymal stem cells into bladder urothelium. J Urol 2008; 180:1778-83. [PMID: 18721942 PMCID: PMC4802964 DOI: 10.1016/j.juro.2008.04.076] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Indexed: 12/21/2022]
Abstract
PURPOSE We have previously reported that embryonic rat bladder mesenchyma has the appropriate inductive signals to direct pluripotent mouse embryonic stem cells toward endodermal derived urothelium and develop mature bladder tissue. We determined whether nonembryonic stem cells, specifically bone marrow derived mesenchymal stem cells, could serve as a source of pluripotent or multipotent progenitor cells. MATERIALS AND METHODS Epithelium was separated from the mesenchymal shells of embryonic day 14 rat bladders. Mesenchymal stem cells were isolated from mouse femoral and tibial bone marrow. Heterospecific recombinant xenografts were created by combining the embryonic rat bladder mesenchyma shells with mesenchymal stem cells and grafting them into the renal subcapsular space of athymic nude mice. Grafts were harvested at time points of up to 42 days and stained for urothelial and stromal differentiation. RESULTS Histological examination of xenografts comprising mouse mesenchymal stem cells and rat embryonic rat bladder mesenchyma yielded mature bladder structures showing normal microscopic architecture as well as proteins confirming functional characteristics. Specifically the induced urothelium expressed uroplakin, a highly selective marker of urothelial differentiation. These differentiated bladder structures demonstrated appropriate alpha-smooth muscle actin staining. Finally, Hoechst staining of the xenografts revealed nuclear architecture consistent with a mouse mesenchymal stem cell origin of the urothelium, supporting differentiated development of these cells. CONCLUSIONS In the appropriate signaling environment bone marrow derived mesenchymal stem cells can undergo directed differentiation toward endodermal derived urothelium and develop into mature bladder tissue in a tissue recombination model. This model serves as an important tool for the study of bladder development with long-term application toward cell replacement therapies in the future.
Collapse
Affiliation(s)
- Govindaraj Anumanthan
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - John H. Makari
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - Lindsey Honea
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - John C. Thomas
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - Marcia L. Wills
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - Neil A. Bhowmick
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - Mark C. Adams
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - Simon W. Hayward
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - Robert J. Matusik
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - John W. Brock
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - John C. Pope
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| |
Collapse
|
48
|
Zhang Y, McNeill E, Tian H, Soker S, Andersson KE, Yoo JJ, Atala A. Urine derived cells are a potential source for urological tissue reconstruction. J Urol 2008; 180:2226-33. [PMID: 18804817 DOI: 10.1016/j.juro.2008.07.023] [Citation(s) in RCA: 249] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2008] [Indexed: 11/29/2022]
Abstract
PURPOSE Contemporary approaches to tissue engineering and cell therapy for urinary tract reconstruction require invasive tissue biopsies to obtain autologous cells. However, these procedures are associated with potential complications. We determined whether the cells present in urine have characteristics of normal bladder cells and investigated their potential uses for urological reconstructive procedures. MATERIALS AND METHODS A total of 55 urine samples were collected from 15 healthy individuals and 8 patients with vesicoureteral reflux. Urine derived cells were isolated, expanded and tested for progenitor and differentiated cell specific markers using flow cytometry, immunofluorescence and Western immunoblotting. The chromosomal stability of cultured urine derived cells was determined by karyotype analysis. RESULTS Clones were successfully established from primary cultures of urine derived cells. Isolated cells showed 3 phenotypes, including fully differentiated, differentiating and progenitor-like cells. Some urine derived cells stained positive for the surface markers c-Kit, SSEA4, CD105, CD73, CD91, CD133 and CD44. Two to 7 cells per 100 ml urine were multipoint progenitors that could expand extensively in culture. Single progenitor cells had the ability to differentiate into the cell lineages expressing urothelial, smooth muscle, endothelial and interstitial cell markers. The expression of lineage markers was characterized by Western blot and immunofluorescence analysis. Urine derived cells also maintained a normal karyotype after serial culture. CONCLUSIONS A subpopulation of cells isolated from urine had progenitor cell features and the potential to differentiate into several bladder cell lineages. Urine derived cells could serve as an alternative cell source for urinary tract tissue engineering and reconstruction.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Institute for Regenerative Medicine, School of Medicine, Wake Forest University, Winston-Salem, North Carolina 27157, USA.
| | | | | | | | | | | | | |
Collapse
|
49
|
Hodges SJ, Atala A. Regenerative medicine and the neurogenic bladder. CURRENT BLADDER DYSFUNCTION REPORTS 2008. [DOI: 10.1007/s11884-008-0011-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
50
|
Campbell GR, Turnbull G, Xiang L, Haines M, Armstrong S, Rolfe BE, Campbell JH. The peritoneal cavity as a bioreactor for tissue engineering visceral organs: bladder, uterus and vas deferens. J Tissue Eng Regen Med 2008; 2:50-60. [DOI: 10.1002/term.66] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|