151
|
3D Co-Culture with Vascular Cells Supports Long-Term Hepatocyte Phenotype and Function In Vitro. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2018. [DOI: 10.1007/s40883-018-0046-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
152
|
Lebaschi A, Nakagawa Y, Wada S, Cong GT, Rodeo SA. Tissue-specific endothelial cells: a promising approach for augmentation of soft tissue repair in orthopedics. Ann N Y Acad Sci 2018; 1410:44-56. [PMID: 29265420 DOI: 10.1111/nyas.13575] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 11/12/2017] [Accepted: 11/13/2017] [Indexed: 12/13/2022]
Abstract
Biologics are playing an increasingly significant role in the practice of modern medicine and surgery in general and orthopedics in particular. Cell-based approaches are among the most important and widely used modalities in orthopedic biologics, with mesenchymal stem cells and other multi/pluripotent cells undergoing evaluation in numerous preclinical and clinical studies. On the other hand, fully differentiated endothelial cells (ECs) have been found to perform critical roles in homeostasis of visceral tissues through production of an adaptive panel of so-called "angiocrine factors." This newly discovered function of ECs renders them excellent candidates for novel approaches in cell-based biologics. Here, we present a review of the role of ECs and angiocrine factors in some visceral tissues, followed by an overview of current cell-based approaches and a discussion of the potential applications of ECs in soft tissue repair.
Collapse
Affiliation(s)
- Amir Lebaschi
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| | - Yusuke Nakagawa
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| | - Susumu Wada
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| | - Guang-Ting Cong
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| | - Scott A Rodeo
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York.,Sports Medicine and Shoulder Service, Hospital for Special Surgery, New York, New York
| |
Collapse
|
153
|
Crawford JM, Bioulac-Sage P, Hytiroglou P. Structure, Function, and Responses to Injury. MACSWEEN'S PATHOLOGY OF THE LIVER 2018:1-87. [DOI: 10.1016/b978-0-7020-6697-9.00001-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
154
|
van Dam PJ, van der Stok EP, Teuwen LA, Van den Eynden GG, Illemann M, Frentzas S, Majeed AW, Eefsen RL, Coebergh van den Braak RRJ, Lazaris A, Fernandez MC, Galjart B, Laerum OD, Rayes R, Grünhagen DJ, Van de paer M, Sucaet Y, Mudhar HS, Schvimer M, Nyström H, Kockx M, Bird NC, Vidal-Vanaclocha F, Metrakos P, Simoneau E, Verhoef C, Dirix LY, Van Laere S, Gao ZH, Brodt P, Reynolds AR, Vermeulen PB. International consensus guidelines for scoring the histopathological growth patterns of liver metastasis. Br J Cancer 2017; 117:1427-1441. [PMID: 28982110 PMCID: PMC5680474 DOI: 10.1038/bjc.2017.334] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 06/12/2017] [Accepted: 08/30/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Liver metastases present with distinct histopathological growth patterns (HGPs), including the desmoplastic, pushing and replacement HGPs and two rarer HGPs. The HGPs are defined owing to the distinct interface between the cancer cells and the adjacent normal liver parenchyma that is present in each pattern and can be scored from standard haematoxylin-and-eosin-stained (H&E) tissue sections. The current study provides consensus guidelines for scoring these HGPs. METHODS Guidelines for defining the HGPs were established by a large international team. To assess the validity of these guidelines, 12 independent observers scored a set of 159 liver metastases and interobserver variability was measured. In an independent cohort of 374 patients with colorectal liver metastases (CRCLM), the impact of HGPs on overall survival after hepatectomy was determined. RESULTS Good-to-excellent correlations (intraclass correlation coefficient >0.5) with the gold standard were obtained for the assessment of the replacement HGP and desmoplastic HGP. Overall survival was significantly superior in the desmoplastic HGP subgroup compared with the replacement or pushing HGP subgroup (P=0.006). CONCLUSIONS The current guidelines allow for reproducible determination of liver metastasis HGPs. As HGPs impact overall survival after surgery for CRCLM, they may serve as a novel biomarker for individualised therapies.
Collapse
Affiliation(s)
- Pieter-Jan van Dam
- Translational Cancer Research Unit, GZA Hospitals (St Augustinus), Wilrijk-Antwerp, Belgium
| | - Eric P van der Stok
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Laure-Anne Teuwen
- Translational Cancer Research Unit, GZA Hospitals (St Augustinus), Wilrijk-Antwerp, Belgium
| | - Gert G Van den Eynden
- Translational Cancer Research Unit, GZA Hospitals (St Augustinus), Wilrijk-Antwerp, Belgium
| | - Martin Illemann
- The Finsen Laboratory, Rigshospitalet/BRIC, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sophia Frentzas
- Tumour Biology Team, Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Ali W Majeed
- Hepatobiliary Surgery, Sheffield Teaching Hospitals, Sheffield, UK
| | - Rikke L Eefsen
- Department of Oncology, Naestved Hospital, Naestved, Denmark
| | | | - Anthoula Lazaris
- Department of Surgery, Cancer Research Program, McGill University Health Centre Research Institute, Montreal, QC, Canada
| | - Maria Celia Fernandez
- Departments of Surgery, Oncology and Medicine, McGill University and the McGill University Health Center Research Institute, Cancer Research Program, Montreal, QC, Canada
| | - Boris Galjart
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Ole Didrik Laerum
- The Finsen Laboratory and Department of Radiation Biology, Copenhagen University Hospital, University of Copenhagen, Denmark
| | - Roni Rayes
- Departments of Surgery, Oncology and Medicine, McGill University and the McGill University Health Center Research Institute, Cancer Research Program, Montreal, QC, Canada
| | - Dirk J Grünhagen
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Michelle Van de paer
- Translational Cancer Research Unit, GZA Hospitals (St Augustinus), Wilrijk-Antwerp, Belgium
- HistoGeneX, Sint-Bavostraat 78-80, Antwerp 2610, Belgium
| | - Yves Sucaet
- Department of Pathology, Faculty of Medicine, Vrije Universiteit Brussel, Brussels, Belgium
- Pathomation, Berchem, Belgium
| | | | - Michael Schvimer
- Institute of Pathology, Sheba Medical Center, Tel Hashomer, Israel
| | - Hanna Nyström
- Department of Surgery, Department of Surgical and Perioperative Sciences, Umeå University, Umeå, Sweden
| | - Mark Kockx
- HistoGeneX, Sint-Bavostraat 78-80, Antwerp 2610, Belgium
| | - Nigel C Bird
- Department of Oncology & Metabolism, University of Sheffield, Sheffield, UK
| | | | - Peter Metrakos
- Department of Surgery, Cancer Research Program, McGill University Health Centre Research Institute, Montreal, QC, Canada
| | - Eve Simoneau
- Department of Surgery, Cancer Research Program, McGill University Health Centre Research Institute, Montreal, QC, Canada
| | - Cornelis Verhoef
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Luc Y Dirix
- Translational Cancer Research Unit, GZA Hospitals (St Augustinus), Wilrijk-Antwerp, Belgium
| | - Steven Van Laere
- Translational Cancer Research Unit, GZA Hospitals (St Augustinus), Wilrijk-Antwerp, Belgium
| | - Zu-hua Gao
- Department of Pathology and Oncology, McGill University, Montreal, QC, Canada
| | - Pnina Brodt
- Departments of Surgery, Oncology and Medicine, McGill University and the McGill University Health Center Research Institute, Cancer Research Program, Montreal, QC, Canada
| | - Andrew R Reynolds
- Tumour Biology Team, Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- Early Clinical Development, Innovative Medicines and Early Development, AstraZeneca, Cambridge, UK
| | - Peter B Vermeulen
- Translational Cancer Research Unit, GZA Hospitals (St Augustinus), Wilrijk-Antwerp, Belgium
- Tumour Biology Team, Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- HistoGeneX, Sint-Bavostraat 78-80, Antwerp 2610, Belgium
| |
Collapse
|
155
|
Kwon HR, Nelson DA, DeSantis KA, Morrissey JM, Larsen M. Endothelial cell regulation of salivary gland epithelial patterning. Development 2017; 144:211-220. [PMID: 28096213 DOI: 10.1242/dev.142497] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 11/10/2016] [Indexed: 12/19/2022]
Abstract
Perfusion-independent regulation of epithelial pattern formation by the vasculature during organ development and regeneration is of considerable interest for application in restoring organ function. During murine submandibular salivary gland development, the vasculature co-develops with the epithelium during branching morphogenesis; however, it is not known whether the vasculature has instructive effects on the epithelium. Using pharmacological inhibitors and siRNA knockdown in embryonic organ explants, we determined that VEGFR2-dependent signaling is required for salivary gland epithelial patterning. To test directly for a requirement for endothelial cells in instructive epithelial patterning, we developed a novel ex vivo cell fractionation/reconstitution assay. Immuno-depletion of CD31+ endothelial cells in this assay confirmed a requirement for endothelial cells in epithelial patterning of the gland. Depletion of endothelial cells or inhibition of VEGFR2 signaling in organ explants caused an aberrant increase in cells expressing the ductal proteins K19 and K7, with a reduction in Kit+ progenitor cells in the endbuds of reconstituted glands. Addition of exogenous endothelial cells to reconstituted glands restored epithelial patterning, as did supplementation with the endothelial cell-regulated mesenchymal factors IGFBP2 and IGFBP3. Our results demonstrate that endothelial cells promote expansion of Kit+ progenitor cells and suppress premature ductal differentiation in early developing embryonic submandibular salivary gland buds.
Collapse
Affiliation(s)
- Hae Ryong Kwon
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA.,Graduate Program in Molecular, Cellular, Developmental, and Neural Biology, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
| | - Deirdre A Nelson
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
| | - Kara A DeSantis
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA.,Graduate Program in Molecular, Cellular, Developmental, and Neural Biology, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
| | - Jennifer M Morrissey
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
| | - Melinda Larsen
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
| |
Collapse
|
156
|
Kedem A, Aelion-Brauer A, Guo P, Wen D, Ding BS, Lis R, Cheng D, Sandler VM, Rafii S, Rosenwaks Z. Activated ovarian endothelial cells promote early follicular development and survival. J Ovarian Res 2017; 10:64. [PMID: 28927438 PMCID: PMC5605973 DOI: 10.1186/s13048-017-0354-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/31/2017] [Indexed: 12/30/2022] Open
Abstract
Background New data suggests that endothelial cells (ECs) elaborate essential “angiocrine factors”. The aim of this study is to investigate the role of activated ovarian endothelial cells in early in-vitro follicular development. Methods Mouse ovarian ECs were isolated using magnetic cell sorting or by FACS and cultured in serum free media. After a constitutive activation of the Akt pathway was initiated, early follicles (50–150 um) were mechanically isolated from 8-day-old mice and co-cultured with these activated ovarian endothelial cells (AOEC) (n = 32), gel (n = 24) or within matrigel (n = 27) in serum free media for 14 days. Follicular growth, survival and function were assessed. Results After 6 passages, flow cytometry showed 93% of cells grown in serum-free culture were VE-cadherin positive, CD-31 positive and CD 45 negative, matching the known EC profile. Beginning on day 4 of culture, we observed significantly higher follicular and oocyte growth rates in follicles co-cultured with AOECs compared with follicles on gel or matrigel. After 14 days of culture, 73% of primary follicles and 83% of secondary follicles co-cultured with AOEC survived, whereas the majority of follicles cultured on gel or matrigel underwent atresia. Conclusions This is the first report of successful isolation and culture of ovarian ECs. We suggest that co-culture with activated ovarian ECs promotes early follicular development and survival. This model is a novel platform for the in vitro maturation of early follicles and for the future exploration of endothelial-follicular communication. Capsule In vitro development of early follicles necessitates a complex interplay of growth factors and signals required for development. Endothelial cells (ECs) may elaborate essential “angiocrine factors” involved in organ regeneration. We demonstrate that co-culture with ovarian ECs enables culture of primary and early secondary mouse ovarian follicles.
Collapse
Affiliation(s)
- Alon Kedem
- The Center for Reproductive Medicine and Infertility, Weill Cornell Medical College, New York, NY, 10021, USA. .,Ansary Stem Cell Center for Regenerative Medicine at Weill Medical College of Cornell University, New York, USA.
| | - Anate Aelion-Brauer
- The Center for Reproductive Medicine and Infertility, Weill Cornell Medical College, New York, NY, 10021, USA.
| | - Peipei Guo
- Ansary Stem Cell Center for Regenerative Medicine at Weill Medical College of Cornell University, New York, USA
| | - Duancheng Wen
- Ansary Stem Cell Center for Regenerative Medicine at Weill Medical College of Cornell University, New York, USA
| | - Bi-Sen Ding
- Ansary Stem Cell Center for Regenerative Medicine at Weill Medical College of Cornell University, New York, USA
| | - Raphael Lis
- Ansary Stem Cell Center for Regenerative Medicine at Weill Medical College of Cornell University, New York, USA
| | - Du Cheng
- Ansary Stem Cell Center for Regenerative Medicine at Weill Medical College of Cornell University, New York, USA
| | - Vladislav M Sandler
- Ansary Stem Cell Center for Regenerative Medicine at Weill Medical College of Cornell University, New York, USA
| | - Shahin Rafii
- Ansary Stem Cell Center for Regenerative Medicine at Weill Medical College of Cornell University, New York, USA
| | - Zev Rosenwaks
- The Center for Reproductive Medicine and Infertility, Weill Cornell Medical College, New York, NY, 10021, USA.,Ansary Stem Cell Center for Regenerative Medicine at Weill Medical College of Cornell University, New York, USA
| |
Collapse
|
157
|
McCauley HA, Wells JM. Pluripotent stem cell-derived organoids: using principles of developmental biology to grow human tissues in a dish. Development 2017; 144:958-962. [PMID: 28292841 DOI: 10.1242/dev.140731] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pluripotent stem cell (PSC)-derived organoids are miniature, three-dimensional human tissues generated by the application of developmental biological principles to PSCs in vitro The approach to generate organoids uses a combination of directed differentiation, morphogenetic processes, and the intrinsically driven self-assembly of cells that mimics organogenesis in the developing embryo. The resulting organoids have remarkable cell type complexity, architecture and function similar to their in vivo counterparts. In the past five years, human PSC-derived organoids with components of all three germ layers have been generated, resulting in the establishment of a new human model system. Here, and in the accompanying poster, we provide an overview of how principles of developmental biology have been essential for generating human organoids in vitro, and how organoids are now being used as a primary research tool to investigate human developmental biology.
Collapse
Affiliation(s)
- Heather A McCauley
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - James M Wells
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA .,Division of Endocrinology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| |
Collapse
|
158
|
Asai A, Aihara E, Watson C, Mourya R, Mizuochi T, Shivakumar P, Phelan K, Mayhew C, Helmrath M, Takebe T, Wells J, Bezerra JA. Paracrine signals regulate human liver organoid maturation from induced pluripotent stem cells. Development 2017; 144:1056-1064. [PMID: 28275009 DOI: 10.1242/dev.142794] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 02/01/2017] [Indexed: 12/17/2022]
Abstract
A self-organizing organoid model provides a new approach to study the mechanism of human liver organogenesis. Previous animal models documented that simultaneous paracrine signaling and cell-to-cell surface contact regulate hepatocyte differentiation. To dissect the relative contributions of the paracrine effects, we first established a liver organoid using human induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs) and human umbilical vein endothelial cells (HUVECs) as previously reported. Time-lapse imaging showed that hepatic-specified endoderm iPSCs (HE-iPSCs) self-assembled into three-dimensional organoids, resulting in hepatic gene induction. Progressive differentiation was demonstrated by hepatic protein production after in vivo organoid transplantation. To assess the paracrine contributions, we employed a Transwell system in which HE-iPSCs were separately co-cultured with MSCs and/or HUVECs. Although the three-dimensional structure did not form, their soluble factors induced a hepatocyte-like phenotype in HE-iPSCs, resulting in the expression of bile salt export pump. In conclusion, the mesoderm-derived paracrine signals promote hepatocyte maturation in liver organoids, but organoid self-organization requires cell-to-cell surface contact. Our in vitro model demonstrates a novel approach to identify developmental paracrine signals regulating the differentiation of human hepatocytes.
Collapse
Affiliation(s)
- Akihiro Asai
- Pediatric Liver Care Center, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Eitaro Aihara
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Carey Watson
- Division of Pediatric Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Reena Mourya
- Pediatric Liver Care Center, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Tatsuki Mizuochi
- Pediatric Liver Care Center, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Pranavkumar Shivakumar
- Pediatric Liver Care Center, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Kieran Phelan
- Pediatric Liver Care Center, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Christopher Mayhew
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michael Helmrath
- Division of Pediatric Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Takanori Takebe
- Department of Regenerative Medicine, Yokohama City University, Yokohama, Kanagawa 236-0004, Japan
| | - James Wells
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jorge A Bezerra
- Pediatric Liver Care Center, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
159
|
Wang JD, An Y, Zhang JS, Wan XH, Jonas JB, Xu L, Zhang W. Human bone marrow mesenchymal stem cells for retinal vascular injury. Acta Ophthalmol 2017; 95:e453-e461. [PMID: 27807930 DOI: 10.1111/aos.13154] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/10/2016] [Indexed: 11/30/2022]
Abstract
PURPOSE To examine the potential of intravitreally implanted human bone marrow-derived mesenchymal stem cells (BMSCs) to affect vascular repair and the blood-retina barrier in mice and rats with oxygen-induced retinopathy, diabetic retinopathy or retinal ischaemia-reperfusion damage. METHODS Three study groups (oxygen-induced retinopathy group: 18 C57BL/6J mice; diabetic retinopathy group: 15 rats; retinal ischaemia-reperfusion model: 18 rats) received BMSCs injected intravitreally. Control groups (oxygen-induced retinopathy group: 12 C57BL/6J mice; diabetic retinopathy group: 15 rats; retinal ischaemia-reperfusion model: 18 rats) received an intravitreal injection of phosphate-buffered saline. We applied immunohistological techniques to measure retinal vascularization, spectroscopic measurements of intraretinally extravasated fluorescein-conjugated dextran to quantify the blood-retina barrier breakdown, and histomorphometry to assess retinal thickness and retinal ganglion cell count. RESULTS In the oxygen-induced retinopathy model, the study group with intravitreally injected BMSCs as compared with the control group showed a significantly (p = 0.001) smaller area of retinal neovascularization. In the diabetic retinopathy model, study group and control group did not differ significantly in the amount of intraretinally extravasated dextran. In the retinal ischaemia-reperfusion model, on the 7th day after retina injury, the retina was significantly thicker in the study group than in the control group (p = 0.02), with no significant difference in the retinal ganglion cell count (p = 0.36). CONCLUSIONS Intravitreally implanted human BMSCs were associated with a reduced retinal neovascularization in the oxygen-induced retinopathy model and with a potentially cell preserving effect in the retinal ischaemia-reperfusion model. Intravitreal BMSCs may be of potential interest for the therapy of retinal vascular disorders.
Collapse
Affiliation(s)
- Jin-Da Wang
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
| | - Ying An
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
| | - Jing-Shang Zhang
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
| | - Xiu-Hua Wan
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
| | - Jost B. Jonas
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
- Department of Ophthalmology; Medical Faculty Mannheim of the Ruprecht-Karls-University Heidelberg; Seegartenklinik; Heidelberg Germany
| | - Liang Xu
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
| | - Wei Zhang
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
| |
Collapse
|
160
|
Wang JD, An Y, Zhang JS, Wan XH, Zhang W, Lanza R, Lu SJ, Jonas JB, Xu L. Retinal vascular injuries and intravitreal human embryonic stem cell-derived haemangioblasts. Acta Ophthalmol 2017. [PMID: 28636206 DOI: 10.1111/aos.13477] [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: 01/17/2023]
Abstract
OBJECTIVE To investigate whether intravitreally applied haemangioblasts (HB) derived from human embryonic stem cells (hESCs) are helpful for the repair of vascular damage caused in animals by an oxygen-induced retinopathy (OIR), by an induced diabetic retinopathy (DR) or by an induced retinal ischaemia with subsequent reperfusion. METHODS Human embryonic stem cell-derived HBs were transplanted intravitreally into C57BL/6J mice (OIR model), into male Wistar rats with an induced DR and into male Wistar rats undergoing induced retinal ischaemia with subsequent reperfusion. Control groups of animals received an intravitreal injection of endothelial cells (ECs) or phosphate-buffered saline (PBS). We examined the vasculature integrity in the mice with OIR, the blood-retina barrier in the rats with induced DR, and retinal thickness and retinal ganglion cell density in retina flat mounts of the rats with the retinal ischaemic-reperfusion retinopathy. RESULTS In the OIR model, the study group versus control groups showed a significantly (p < 0.001) smaller retinal avascular area [5.1 ± 2.7%;n = 18 animals versus 12.2 ± 2.8% (PBS group; n = 10 animals) and versus 11.8 ± 3.7% (EC group; n = 8 animals)] and less retinal neovascularization [6.3 ± 2.5%;n = 18 versus 15.2 ± 6.3% (n = 10; PBS group) and versus 15.8 ± 3.3% (n = 8; EC group)]. On retinal flat mounts, hESC-HBs were integrated into damaged retinal vessels and stained positive for PECAM (CD31) as EC marker. In the DR model, the study group versus the EC control group showed a significantly (p = 0.001) better blood-retina barrier function as measured at 2 days after the intravitreal injections [study group: 20.2 ± 12.8 μl/(g × hr); n = 6; versus EC control group: 52.9 ± 9.9 μl/(g × hr; n = 6)]. In the retinal ischaemia-reperfusion model, the groups did not differ significantly in retinal thickness and retinal ganglion cell density at 2, 5 and 7 days after baseline. CONCLUSION By integrating into damaged retinal vessels and differentiating into ECs, intravitreally administered hESC-HBs may have partially repaired a retinal vascular injury caused by OIR model and DR.
Collapse
Affiliation(s)
- Jin-Da Wang
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing Tongren Hospital of Capital Medical University; Beijing China
| | - Ying An
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing Tongren Hospital of Capital Medical University; Beijing China
| | - Jing-Shang Zhang
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing Tongren Hospital of Capital Medical University; Beijing China
| | - Xiu-Hua Wan
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing Tongren Hospital of Capital Medical University; Beijing China
| | - Wei Zhang
- University of North Texas Health Science Center; Fort Worth Texas USA
| | - Robert Lanza
- Ocata Therapeutics; Marlborough Massachusetts USA
| | - Shi-Jiang Lu
- Ocata Therapeutics; Marlborough Massachusetts USA
| | - Jost B. Jonas
- Department of Ophthalmology; Medical Faculty Mannheim of the Ruprecht-Karls-University Heidelberg; Mannheim Germany
| | - Liang Xu
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing Tongren Hospital of Capital Medical University; Beijing China
| |
Collapse
|
161
|
Carballo CB, Lebaschi A, Rodeo SA. Cell-based approaches for augmentation of tendon repair. TECHNIQUES IN SHOULDER & ELBOW SURGERY 2017; 18:e6-e14. [PMID: 29276433 PMCID: PMC5737795 DOI: 10.1097/bte.0000000000000132] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell-based approaches are among the principal interventions in orthobiologics to improve tendon and ligament healing and to combat degenerative processes. The number of options available for investigation are expanding rapidly and investigators have an increasing number of cell types to choose from for research purposes. However, in part due to the current regulatory environment, the list of available cells at clinicians' disposal for therapeutic purposes is still rather limited. In this review, we present an overview of the main cellular categories in current use. Notable recent developments in cell-based approaches include the introduction of diverse sources of mesenchymal stem cells, pluripotent cells of extra-embryonic origin, and the emerging popularity of fully differentiated cells such as tenocytes and endothelial cells. Delivery strategies are discussed and a succinct discussion of the current regulatory environment in the United States is presented.
Collapse
Affiliation(s)
- Camila B Carballo
- Laboratory for Joint Tissue Repair and Regeneration, Orthopedic Soft Tissue Research Program, Hospital for Special Surgery
| | - Amir Lebaschi
- Laboratory for Joint Tissue Repair and Regeneration, Orthopedic Soft Tissue Research Program, Hospital for Special Surgery
| | - Scott A Rodeo
- Laboratory for Joint Tissue Repair and Regeneration, Orthopedic Soft Tissue Research Program, Hospital for Special Surgery
| |
Collapse
|
162
|
Yao J, Guihard PJ, Wu X, Blazquez-Medela AM, Spencer MJ, Jumabay M, Tontonoz P, Fogelman AM, Boström KI, Yao Y. Vascular endothelium plays a key role in directing pulmonary epithelial cell differentiation. J Cell Biol 2017; 216:3369-3385. [PMID: 28838957 PMCID: PMC5626536 DOI: 10.1083/jcb.201612122] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/26/2017] [Accepted: 07/12/2017] [Indexed: 12/19/2022] Open
Abstract
J. Yao et al. demonstrate that loss of MGP, a BMP inhibitor, causes abnormal hepatic differentiation in lungs. They find that interactions between endothelium and epithelium separate pulmonary from hepatic differentiation during development. Lack of MGP triggers hepatic differentiation in the pulmonary epithelium, as regulated by the endothelium. The vascular endothelium is critical for induction of appropriate lineage differentiation in organogenesis. In this study, we report that dysfunctional pulmonary endothelium, resulting from the loss of matrix Gla protein (MGP), causes ectopic hepatic differentiation in the pulmonary epithelium. We demonstrate uncontrolled induction of the hepatic growth factor (HGF) caused by dysregulated cross talk between pulmonary endothelium and epithelium in Mgp-null lungs. Elevated HGF induced hepatocyte nuclear factor 4 α (Hnf4a), which competed with NK2 homeobox 1 (Nkx2.1) for binding to forkhead box A2 (Foxa2) to drive hepatic differentiation in Mgp-null airway progenitor cells. Limiting endothelial HGF reduced Hnf4a, abolished interference of Hnf4a with Foxa2, and reduced hepatic differentiation in Mgp-null lungs. Together, our results suggest that endothelial–epithelial interactions, maintained by MGP, are essential in pulmonary cell differentiation.
Collapse
Affiliation(s)
- Jiayi Yao
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Pierre J Guihard
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Xiuju Wu
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Ana M Blazquez-Medela
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Melissa J Spencer
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Medet Jumabay
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA.,Department of Pathology and Laboratory Medicine, Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA
| | - Alan M Fogelman
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Kristina I Boström
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA .,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA
| | - Yucheng Yao
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA .,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
163
|
Hikspoors JPJM, Peeters MMJP, Mekonen HK, Kruepunga N, Mommen GMC, Cornillie P, Köhler SE, Lamers WH. The fate of the vitelline and umbilical veins during the development of the human liver. J Anat 2017; 231:718-735. [PMID: 28786203 DOI: 10.1111/joa.12671] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2017] [Indexed: 01/15/2023] Open
Abstract
Differentiation of endodermal cells into hepatoblasts is well studied, but the remodeling of the vitelline and umbilical veins during liver development is less well understood. We compared human embryos between 3 and 10 weeks of development with pig and mouse embryos at comparable stages, and used Amira 3D reconstruction and Cinema 4D remodeling software for visualization. The vitelline and umbilical veins enter the systemic venous sinus on each side via a common entrance, the hepatocardiac channel. During expansion into the transverse septum at Carnegie Stage (CS)12 the liver bud develops as two dorsolateral lobes or 'wings' and a single ventromedial lobe, with the liver hilum at the intersection of these lobes. The dorsolateral lobes each engulf a vitelline vein during CS13 and the ventromedial lobe both umbilical veins during CS14, but both venous systems remain temporarily identifiable inside the liver. The dominance of the left-sided umbilical vein and the rightward repositioning of the sinuatrial junction cause de novo development of left-to-right shunts between the left umbilical vein in the liver hilum and the right hepatocardiac channel (venous duct) and the right vitelline vein (portal sinus), respectively. Once these shunts have formed, portal branches develop from the intrahepatic portions of the portal vein on the right side and the umbilical vein on the left side. The gall bladder is a reliable marker for this hepatic vascular midline. We found no evidence for large-scale fragmentation of embryonic veins as claimed by the 'vestigial' theory. Instead and in agreement with the 'lineage' theory, the vitelline and umbilical veins remained temporally identifiable inside the liver after being engulfed by hepatoblasts. In agreement with the 'hemodynamic' theory, the left-right shunts develop de novo.
Collapse
Affiliation(s)
- Jill P J M Hikspoors
- Department of Anatomy & Embryology, Maastricht University, Maastricht, The Netherlands
| | - Mathijs M J P Peeters
- Department of Anatomy & Embryology, Maastricht University, Maastricht, The Netherlands
| | - Hayelom K Mekonen
- Department of Anatomy & Embryology, Maastricht University, Maastricht, The Netherlands
| | - Nutmethee Kruepunga
- Department of Anatomy & Embryology, Maastricht University, Maastricht, The Netherlands
| | - Greet M C Mommen
- Department of Anatomy & Embryology, Maastricht University, Maastricht, The Netherlands
| | - Pieter Cornillie
- Department of Morphology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - S Eleonore Köhler
- Department of Anatomy & Embryology, Maastricht University, Maastricht, The Netherlands.,NUTRIM Research School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Wouter H Lamers
- Department of Anatomy & Embryology, Maastricht University, Maastricht, The Netherlands.,Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
164
|
Effects of Co-Culture Media on Hepatic Differentiation of hiPSC with or without HUVEC Co-Culture. Int J Mol Sci 2017; 18:ijms18081724. [PMID: 28783133 PMCID: PMC5578114 DOI: 10.3390/ijms18081724] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 07/24/2017] [Accepted: 08/02/2017] [Indexed: 12/14/2022] Open
Abstract
The derivation of hepatocytes from human induced pluripotent stem cells (hiPSC) is of great interest for applications in pharmacological research. However, full maturation of hiPSC-derived hepatocytes has not yet been achieved in vitro. To improve hepatic differentiation, co-cultivation of hiPSC with human umbilical vein endothelial cells (HUVEC) during hepatic differentiation was investigated in this study. In the first step, different culture media variations based on hepatocyte culture medium (HCM) were tested in HUVEC mono-cultures to establish a suitable culture medium for co-culture experiments. Based on the results, two media variants were selected to differentiate hiPSC-derived definitive endodermal (DE) cells into mature hepatocytes with or without HUVEC addition. DE cells differentiated in mono-cultures in the presence of those media variants showed a significant increase (p < 0.05) in secretion of α-fetoprotein and in activities of cytochrome P450 (CYP) isoenzymes CYP2B6 and CYP3A4 as compared with cells differentiated in unmodified HCM used as control. Co-cultivation with HUVEC did not further improve the differentiation outcome. Thus, it can be concluded that the effect of the used medium outweighed the effect of HUVEC co-culture, emphasizing the importance of the culture medium composition for hiPSC differentiation.
Collapse
|
165
|
Augustin HG, Koh GY. Organotypic vasculature: From descriptive heterogeneity to functional pathophysiology. Science 2017; 357:science.aal2379. [DOI: 10.1126/science.aal2379] [Citation(s) in RCA: 351] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
166
|
Tanimizu N, Mitaka T. Epithelial Morphogenesis during Liver Development. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a027862. [PMID: 28213465 DOI: 10.1101/cshperspect.a027862] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tissue stem/progenitor cells supply multiple types of epithelial cells that eventually acquire specialized functions during organ development. In addition, three-dimensional (3D) tissue structures need to be established for organs to perform their physiological functions. The liver contains two types of epithelial cells, namely, hepatocytes and cholangiocytes, which are derived from hepatoblasts, fetal liver stem/progenitor cells (LPCs), in mid-gestation. Hepatocytes performing many metabolic reactions form cord-like structures, whereas cholangiocytes, biliary epithelial cells, form tubular structures called intrahepatic bile ducts. Analyses for human genetic diseases and mutant mice have identified crucial molecules for liver organogenesis. Functions of those molecules can be examined in in vitro culture systems where LPCs are induced to differentiate into hepatocytes or cholangiocytes. Recent technical advances have revealed 3D epithelial morphogenesis during liver organogenesis. Therefore, the liver is a good model to understand how tissue stem/progenitor cells differentiate and establish 3D tissue architectures during organ development.
Collapse
Affiliation(s)
- Naoki Tanimizu
- Department of Tissue Development and Regeneration, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo 060-8556, Japan
| | - Toshihiro Mitaka
- Department of Tissue Development and Regeneration, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo 060-8556, Japan
| |
Collapse
|
167
|
Future Challenges in the Generation of Hepatocyte-Like Cells From Human Pluripotent Stem Cells. CURRENT PATHOBIOLOGY REPORTS 2017. [DOI: 10.1007/s40139-017-0150-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
168
|
Koui Y, Kido T, Ito T, Oyama H, Chen SW, Katou Y, Shirahige K, Miyajima A. An In Vitro Human Liver Model by iPSC-Derived Parenchymal and Non-parenchymal Cells. Stem Cell Reports 2017; 9:490-498. [PMID: 28757162 PMCID: PMC5549957 DOI: 10.1016/j.stemcr.2017.06.010] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 02/05/2023] Open
Abstract
During liver development, hepatoblasts and liver non-parenchymal cells (NPCs) such as liver sinusoidal endothelial cells (LSECs) and hepatic stellate cells (HSCs) constitute the liver bud where they proliferate and differentiate. Accordingly, we reasoned that liver NPCs would support the maturation of hepatocytes derived from human induced pluripotent stem cells (hiPSCs), which usually exhibit limited functions. We found that the transforming growth factor β and Rho signaling pathways, respectively, regulated the proliferation and maturation of LSEC and HSC progenitors isolated from mouse fetal livers. Based on these results, we have established culture systems to generate LSECs and HSCs from hiPSCs. These hiPSC-derived NPCs exhibited distinctive phenotypes and promoted self-renewal of hiPSC-derived liver progenitor cells (LPCs) over the long term in the two-dimensional culture system without exogenous cytokines and hepatic maturation of hiPSC-derived LPCs. Thus, a functional human liver model can be constructed in vitro from the LPCs, LSECs, and HSCs derived from hiPSCs.
Collapse
Affiliation(s)
- Yuta Koui
- Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo 113-0032, Japan
| | - Taketomo Kido
- Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo 113-0032, Japan.
| | - Toshimasa Ito
- Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo 113-0032, Japan
| | - Hiroki Oyama
- Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo 113-0032, Japan
| | - Shin-Wei Chen
- Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo 113-0032, Japan
| | - Yuki Katou
- Laboratory of Genome Structure and Function, Research Center for Epigenetic Disease, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo 113-0032, Japan
| | - Katsuhiko Shirahige
- Laboratory of Genome Structure and Function, Research Center for Epigenetic Disease, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo 113-0032, Japan
| | - Atsushi Miyajima
- Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo 113-0032, Japan.
| |
Collapse
|
169
|
History and conceptual developments in vascular biology and angiogenesis research: a personal view. Angiogenesis 2017; 20:463-478. [DOI: 10.1007/s10456-017-9569-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 07/18/2017] [Indexed: 01/05/2023]
|
170
|
Peters EB. Endothelial Progenitor Cells for the Vascularization of Engineered Tissues. TISSUE ENGINEERING PART B-REVIEWS 2017; 24:1-24. [PMID: 28548628 DOI: 10.1089/ten.teb.2017.0127] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Self-assembled microvasculature from cocultures of endothelial cells (ECs) and stromal cells has significantly advanced efforts to vascularize engineered tissues by enhancing perfusion rates in vivo and producing investigative platforms for microvascular morphogenesis in vitro. However, to clinically translate prevascularized constructs, the issue of EC source must be resolved. Endothelial progenitor cells (EPCs) can be noninvasively supplied from the recipient through adult peripheral and umbilical cord blood, as well as derived from induced pluripotent stem cells, alleviating antigenicity issues. EPCs can also differentiate into all tissue endothelium, and have demonstrated potential for therapeutic vascularization. Yet, EPCs are not the standard EC choice to vascularize tissue constructs in vitro. Possible reasons include unresolved issues with EPC identity and characterization, as well as uncertainty in the selection of coculture, scaffold, and culture media combinations that promote EPC microvessel formation. This review addresses these issues through a summary of EPC vascular biology and the effects of tissue engineering design parameters upon EPC microvessel formation. Also included are perspectives to integrate EPCs with emerging technologies to produce functional, organotypic vascularized tissues.
Collapse
Affiliation(s)
- Erica B Peters
- Department of Chemical and Biological Engineering, University of Colorado , Boulder, Colorado
| |
Collapse
|
171
|
Melgar-Lesmes P, Balcells M, Edelman ER. Implantation of healthy matrix-embedded endothelial cells rescues dysfunctional endothelium and ischaemic tissue in liver engraftment. Gut 2017; 66:1297-1305. [PMID: 26851165 PMCID: PMC5288307 DOI: 10.1136/gutjnl-2015-310409] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 01/12/2016] [Accepted: 01/14/2016] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Liver transplantation is limited by ischaemic injury which promotes endothelial cell and hepatocyte dysfunction and eventually organ failure. We sought to understand how endothelial state determines liver recovery after hepatectomy and engraftment. DESIGN Matrix-embedded endothelial cells (MEECs) with retained healthy phenotype or control acellular matrices were implanted in direct contact with the remaining median lobe of donor mice undergoing partial hepatectomy (70%), or in the interface between the remaining median lobe and an autograft or isograft from the left lobe in hepatectomised recipient mice. Hepatic vascular architecture, DNA fragmentation and apoptosis in the median lobe and grafts, serum markers of liver damage and phenotype of macrophage and lymphocyte subsets in the liver after engraftment were analysed 7 days post-op. RESULTS Healthy MEECs create a functional vascular splice in donor and recipient liver after 70% hepatectomy in mouse protecting these livers from ischaemic injury, hepatic congestion and inflammation. Macrophages recruited adjacent to the vascular nodes into the implants switched to an anti-inflammatory and regenerative profile M2. MEECs improved liver function and the rate of liver regeneration and prevented apoptosis in donor liver lobes, autologous grafts and syngeneic engraftment. CONCLUSIONS Implants with healthy endothelial cells rescue liver donor and recipient endothelium and parenchyma from ischaemic injury after major hepatectomy and engraftment. This study highlights endothelial-hepatocyte crosstalk in hepatic repair and provides a promising new approach to improve regenerative medicine outcomes and liver transplantation.
Collapse
Affiliation(s)
- Pedro Melgar-Lesmes
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, US,Corresponding author: Pedro Melgar-Lesmes, PhD, Massachusetts Institute of Technology, Cambridge, Massachusetts; 77 Massachusetts Avenue, Building E25-438. Cambridge, MA 02139. USA. Phone: +1 617-715-2026, FAX: +1 617-253-2514,
| | - Mercedes Balcells
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, US,Bioengineering Department, Institut Químic de Sarrià, Ramon Llull Univ, Barcelona, Spain
| | - Elazer R. Edelman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, US,Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, US
| |
Collapse
|
172
|
Havrilak JA, Melton KR, Shannon JM. Endothelial cells are not required for specification of respiratory progenitors. Dev Biol 2017; 427:93-105. [PMID: 28501476 PMCID: PMC5551037 DOI: 10.1016/j.ydbio.2017.05.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 04/21/2017] [Accepted: 05/08/2017] [Indexed: 11/23/2022]
Abstract
Crosstalk between mesenchymal and epithelial cells influences organogenesis in multiple tissues, such as lung, pancreas, liver, and the nervous system. Lung mesenchyme comprises multiple cell types, however, and precise identification of the mesenchymal cell type(s) that drives early events in lung development remains unknown. Endothelial cells have been shown to be required for some aspects of lung epithelial patterning, lung stem cell differentiation, and regeneration after injury. Furthermore, endothelial cells are involved in early liver and pancreas development. From these observations we hypothesized that endothelial cells might also be required for early specification of the respiratory field and subsequent lung bud initiation. We first blocked VEGF signaling in E8.5 cultured foreguts with small molecule VEGFR inhibitors and found that lung specification and bud formation were unaltered. However, when we examined E9.5 mouse embryos carrying a mutation in the VEGFR Flk-1, which do not develop endothelial cells, we found that respiratory progenitor specification was impeded. Because the E9.5 embryos were substantially smaller than control littermates, suggesting the possibility of developmental delay, we isolated and cultured foreguts from mutant and control embryos on E8.5, when no size differences were apparent. We found that both specification of the respiratory field and lung bud formation occurred in mutant and control explants. These observations were unaffected by the presence or absence of serum. We also observed that hepatic specification and initiation occurred in the absence of endothelial cells, and that expansion of the liver epithelium in culture did not differ between mutant and control explants. Consistent with previously published results, we also found that pancreatic buds were not maintained in cultured foreguts when endothelial cells were absent. Our observations support the conclusion that endothelial cells are not required for early specification of lung progenitors and bud initiation, and that the diminished lung specification seen in E9.5 Flk-/- embryos is likely due to developmental delay resulting from the insufficient delivery of oxygen, nutrients, and other factors in the absence of a vasculature.
Collapse
Affiliation(s)
- Jamie A Havrilak
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, 3333 Burnet Ave, Cincinnati, OH 45229, United States; Section of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229, United States
| | - Kristin R Melton
- Section of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229, United States
| | - John M Shannon
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, 3333 Burnet Ave, Cincinnati, OH 45229, United States; Section of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229, United States.
| |
Collapse
|
173
|
Camp JG, Sekine K, Gerber T, Loeffler-Wirth H, Binder H, Gac M, Kanton S, Kageyama J, Damm G, Seehofer D, Belicova L, Bickle M, Barsacchi R, Okuda R, Yoshizawa E, Kimura M, Ayabe H, Taniguchi H, Takebe T, Treutlein B. Multilineage communication regulates human liver bud development from pluripotency. Nature 2017; 546:533-538. [PMID: 28614297 DOI: 10.1038/nature22796] [Citation(s) in RCA: 407] [Impact Index Per Article: 50.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 04/27/2017] [Indexed: 12/11/2022]
Abstract
Conventional two-dimensional differentiation from pluripotency fails to recapitulate cell interactions occurring during organogenesis. Three-dimensional organoids generate complex organ-like tissues; however, it is unclear how heterotypic interactions affect lineage identity. Here we use single-cell RNA sequencing to reconstruct hepatocyte-like lineage progression from pluripotency in two-dimensional culture. We then derive three-dimensional liver bud organoids by reconstituting hepatic, stromal, and endothelial interactions, and deconstruct heterogeneity during liver bud development. We find that liver bud hepatoblasts diverge from the two-dimensional lineage, and express epithelial migration signatures characteristic of organ budding. We benchmark three-dimensional liver buds against fetal and adult human liver single-cell RNA sequencing data, and find a striking correspondence between the three-dimensional liver bud and fetal liver cells. We use a receptor-ligand pairing analysis and a high-throughput inhibitor assay to interrogate signalling in liver buds, and show that vascular endothelial growth factor (VEGF) crosstalk potentiates endothelial network formation and hepatoblast differentiation. Our molecular dissection reveals interlineage communication regulating organoid development, and illuminates previously inaccessible aspects of human liver development.
Collapse
Affiliation(s)
- J Gray Camp
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, Leipzig 04103, Germany
| | - Keisuke Sekine
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Tobias Gerber
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, Leipzig 04103, Germany
| | - Henry Loeffler-Wirth
- Interdisciplinary Centre for Bioinformatics, Leipzig University, 16 Härtelstrasse, Leipzig 04107, Germany
| | - Hans Binder
- Interdisciplinary Centre for Bioinformatics, Leipzig University, 16 Härtelstrasse, Leipzig 04107, Germany
| | - Malgorzata Gac
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, Leipzig 04103, Germany
| | - Sabina Kanton
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, Leipzig 04103, Germany
| | - Jorge Kageyama
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, Leipzig 04103, Germany
| | - Georg Damm
- Department of Hepatobiliary and Transplantation Surgery, University Hospital of Leipzig, Liebigstrasse 20, Leipzig 04103, Germany.,Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, 55 Philipp-Rosenthal-Strasse, Leipzig 04103, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary and Transplantation Surgery, University Hospital of Leipzig, Liebigstrasse 20, Leipzig 04103, Germany.,Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, 55 Philipp-Rosenthal-Strasse, Leipzig 04103, Germany
| | - Lenka Belicova
- Max Planck Institute of Molecular Cell Biology and Genetics, 108 Pfotenhauerstrasse, Dresden 01307, Germany
| | - Marc Bickle
- Max Planck Institute of Molecular Cell Biology and Genetics, 108 Pfotenhauerstrasse, Dresden 01307, Germany
| | - Rico Barsacchi
- Max Planck Institute of Molecular Cell Biology and Genetics, 108 Pfotenhauerstrasse, Dresden 01307, Germany
| | - Ryo Okuda
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Emi Yoshizawa
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Masaki Kimura
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Hiroaki Ayabe
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Takanori Takebe
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan.,Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, Ohio 45229-3039, USA
| | - Barbara Treutlein
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, Leipzig 04103, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, 108 Pfotenhauerstrasse, Dresden 01307, Germany
| |
Collapse
|
174
|
Guo X, Li W, Ma M, Lu X, Zhang H. Endothelial cell-derived matrix promotes the metabolic functional maturation of hepatocyte via integrin-Src signalling. J Cell Mol Med 2017; 21:2809-2822. [PMID: 28470937 PMCID: PMC5661128 DOI: 10.1111/jcmm.13195] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/19/2017] [Indexed: 01/07/2023] Open
Abstract
The extracellular matrix (ECM) microenvironment is involved in the regulation of hepatocyte phenotype and function. Recently, the cell‐derived extracellular matrix has been proposed to represent the bioactive and biocompatible materials of the native ECM. Here, we show that the endothelial cell‐derived matrix (EC matrix) promotes the metabolic maturation of human adipose stem cell‐derived hepatocyte‐like cells (hASC‐HLCs) through the activation of the transcription factor forkhead box protein A2 (FOXA2) and the nuclear receptors hepatocyte nuclear factor 4 alpha (HNF4α) and pregnane X receptor (PXR). Reducing the fibronectin content in the EC matrix or silencing the expression of α5 integrin in the hASC‐HLCs inhibited the effect of the EC matrix on Src phosphorylation and hepatocyte maturation. The inhibition of Src phosphorylation using the inhibitor PP2 or silencing the expression of Src in hASC‐HLCs also attenuated the up‐regulation of the metabolic function of hASC‐HLCs in a nuclear receptor‐dependent manner. These data elucidate integrin‐Src signalling linking the extrinsic EC matrix signals and metabolic functional maturation of hepatocyte. This study provides a model for studying the interaction between hepatocytes and non‐parenchymal cell‐derived matrix.
Collapse
Affiliation(s)
- Xinyue Guo
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Weihong Li
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Minghui Ma
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Xin Lu
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Haiyan Zhang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| |
Collapse
|
175
|
Hypertrophic scar regression is linked to the occurrence of endothelial dysfunction. PLoS One 2017; 12:e0176681. [PMID: 28472181 PMCID: PMC5417599 DOI: 10.1371/journal.pone.0176681] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/14/2017] [Indexed: 11/19/2022] Open
Abstract
Most microvessels have been shown to become stenosed or completely occluded during hypertrophic scar progression. Here, we examined the morphology of capillary endothelial cells (ECs) and fibroblasts using immunofluorescence staining for CD31 and alpha-smooth muscle actin (α-SMA) and electron microscopy. In addition, ECs and fibroblasts were isolated from scar tissues, and the levels of transforming growth factor beta 1 (TGF-β1), platelet-derived growth factor (PDGF), endothelin 1 (ET-1), vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) were assayed using ELISAs. Furthermore, we assessed cell viability, total collagen production, and cell apoptosis in hypertrophic scar-derived fibroblasts cultured with EC-conditioned medium. Then, anti-TGF-β1, anti-PDGF, anti-ET-1, anti-VEGF, and anti-bFGF neutralising antibodies were individually added to the EC medium to identify which growth factor plays a more important role in inhibiting fibroblasts biology. Our results showed microvessel lumen occlusion and EC atrophy during scar development, particularly in regressive scars (RSs). Additionally, EC growth factor secretion decreased and reached the lowest levels in RSs. Furthermore, based on the culture results, RS EC medium inhibited fibroblast viability and collagen production and induced apoptosis. Moreover, TGF-β1, PDGF, and bFGF played more important roles in these processes than VEGF and ET-1. The endothelial dysfunction occurring in hypertrophic scars contributes to fibroblast inhibition and scar regression, and reduced TGF-β1, PDGF, and bFGF levels play key roles during this process.
Collapse
|
176
|
Koike H, Zhang RR, Ueno Y, Sekine K, Zheng YW, Takebe T, Taniguchi H. Nutritional modulation of mouse and human liver bud growth through a branched-chain amino acid metabolism. Development 2017; 144:1018-1024. [PMID: 28219950 DOI: 10.1242/dev.143032] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 02/13/2017] [Indexed: 12/31/2022]
Abstract
Liver bud progenitors experience a transient amplification during the early organ growth phase, yet the mechanism responsible is not fully understood. Collective evidence highlights the specific requirements in stem cell metabolism for expanding organ progenitors during organogenesis and regeneration. Here, transcriptome analyses show that progenitors of the mouse and human liver bud growth stage specifically express the gene branched chain aminotransferase 1, encoding a known breakdown enzyme of branched-chain amino acids (BCAAs) for energy generation. Global metabolome analysis confirmed the active consumption of BCAAs in the growing liver bud, but not in the later fetal or adult liver. Consistently, maternal dietary restriction of BCAAs during pregnancy significantly abrogated the conceptus liver bud growth capability through a striking defect in hepatic progenitor expansion. Under defined conditions, the supplementation of L-valine specifically among the BCAAs promoted rigorous growth of the human liver bud organoid in culture by selectively amplifying self-renewing bi-potent hepatic progenitor cells. These results highlight a previously underappreciated role of branched-chain amino acid metabolism in regulating mouse and human liver bud growth that can be modulated by maternal nutrition in vivo or cultural supplement in vitro.
Collapse
Affiliation(s)
- Hiroyuki Koike
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Ran-Ran Zhang
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Yasuharu Ueno
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Keisuke Sekine
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Yun-Wen Zheng
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Takanori Takebe
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
- Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa 236-0004, Japan
- PRESTO, Japan Science and Technology Agency, 4-1-8, Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
- Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa 236-0004, Japan
| |
Collapse
|
177
|
Wieland E, Rodriguez-Vita J, Liebler SS, Mogler C, Moll I, Herberich SE, Espinet E, Herpel E, Menuchin A, Chang-Claude J, Hoffmeister M, Gebhardt C, Brenner H, Trumpp A, Siebel CW, Hecker M, Utikal J, Sprinzak D, Fischer A. Endothelial Notch1 Activity Facilitates Metastasis. Cancer Cell 2017; 31:355-367. [PMID: 28238683 DOI: 10.1016/j.ccell.2017.01.007] [Citation(s) in RCA: 234] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 09/28/2016] [Accepted: 01/25/2017] [Indexed: 12/11/2022]
Abstract
Endothelial cells (ECs) provide angiocrine factors orchestrating tumor progression. Here, we show that activated Notch1 receptors (N1ICD) are frequently observed in ECs of human carcinomas and melanoma, and in ECs of the pre-metastatic niche in mice. EC N1ICD expression in melanoma correlated with shorter progression-free survival. Sustained N1ICD activity induced EC senescence, expression of chemokines and the adhesion molecule VCAM1. This promoted neutrophil infiltration, tumor cell (TC) adhesion to the endothelium, intravasation, lung colonization, and postsurgical metastasis. Thus, sustained vascular Notch signaling facilitates metastasis by generating a senescent, pro-inflammatory endothelium. Consequently, treatment with Notch1 or VCAM1-blocking antibodies prevented Notch-driven metastasis, and genetic ablation of EC Notch signaling inhibited peritoneal neutrophil infiltration in an ovarian carcinoma mouse model.
Collapse
Affiliation(s)
- Elfriede Wieland
- Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Juan Rodriguez-Vita
- Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sven S Liebler
- Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Vascular Biology, CBTM, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Carolin Mogler
- Institute of Pathology, Heidelberg University Hospital, Vascular Oncology and Metastasis (A190), German Cancer Research Center, 69120 Heidelberg, Germany
| | - Iris Moll
- Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Stefanie E Herberich
- Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Vascular Biology, CBTM, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Elisa Espinet
- Division of Stem Cells and Cancer (A010), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance and the German Cancer Consortium (DKTK), Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
| | - Esther Herpel
- Tissue Bank of the National Center for Tumor Diseases (NCT) Heidelberg, Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Amitai Menuchin
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Tel-Aviv University, 69978 Tel Aviv, Israel
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology (C020), German Cancer Research Center, 69120 Heidelberg, Germany; Research Group Genetic Cancer Epidemiology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research (C070), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Christoffer Gebhardt
- Clinical Cooperation Unit Dermato-Oncology (G300), German Cancer Research Center (DKFZ), Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, 69120 Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research (C070), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Division of Preventive Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer (A010), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance and the German Cancer Consortium (DKTK), Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
| | - Christian W Siebel
- Department of Discovery Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Markus Hecker
- Department of Cardiovascular Physiology, Heidelberg University and Deutsches Zentrum für Herz-Kreislauf-Forschung e.V. (DZHK), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Jochen Utikal
- Clinical Cooperation Unit Dermato-Oncology (G300), German Cancer Research Center (DKFZ), Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, 69120 Heidelberg, Germany
| | - David Sprinzak
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Tel-Aviv University, 69978 Tel Aviv, Israel
| | - Andreas Fischer
- Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Vascular Biology, CBTM, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; Department of Medicine I and Clinical Chemistry, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| |
Collapse
|
178
|
Caceres PS, Benedicto I, Lehmann GL, Rodriguez-Boulan EJ. Directional Fluid Transport across Organ-Blood Barriers: Physiology and Cell Biology. Cold Spring Harb Perspect Biol 2017; 9:a027847. [PMID: 28003183 PMCID: PMC5334253 DOI: 10.1101/cshperspect.a027847] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Directional fluid flow is an essential process for embryo development as well as for organ and organism homeostasis. Here, we review the diverse structure of various organ-blood barriers, the driving forces, transporters, and polarity mechanisms that regulate fluid transport across them, focusing on kidney-, eye-, and brain-blood barriers. We end by discussing how cross talk between barrier epithelial and endothelial cells, perivascular cells, and basement membrane signaling contribute to generate and maintain organ-blood barriers.
Collapse
Affiliation(s)
- Paulo S Caceres
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| | - Ignacio Benedicto
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| | - Guillermo L Lehmann
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| | - Enrique J Rodriguez-Boulan
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| |
Collapse
|
179
|
CÉLULAS MADRE: FUNDAMENTOS Y REVISIÓN DE LA EXPERIENCIA CLÍNICA EN ENFERMEDADES HEPÁTICAS. REVISTA MÉDICA CLÍNICA LAS CONDES 2017. [DOI: 10.1016/j.rmclc.2017.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
180
|
de Paula F, Teshima THN, Hsieh R, Souza MM, Nico MMS, Lourenco SV. Overview of Human Salivary Glands: Highlights of Morphology and Developing Processes. Anat Rec (Hoboken) 2017; 300:1180-1188. [DOI: 10.1002/ar.23569] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 09/13/2016] [Accepted: 11/01/2016] [Indexed: 01/12/2023]
Affiliation(s)
- Fernanda de Paula
- Department of Dermatology; School of Medicine, University of Sao Paulo; Sao Paulo Brazil
| | | | - Ricardo Hsieh
- Department of Stomatology; School of Dentistry, University of Sao Paulo; Sao Paulo Brazil
| | - Milena Monteiro Souza
- Department of Dermatology; School of Medicine, University of Sao Paulo; Sao Paulo Brazil
| | | | | |
Collapse
|
181
|
Géraud C, Koch PS, Zierow J, Klapproth K, Busch K, Olsavszky V, Leibing T, Demory A, Ulbrich F, Diett M, Singh S, Sticht C, Breitkopf-Heinlein K, Richter K, Karppinen SM, Pihlajaniemi T, Arnold B, Rodewald HR, Augustin HG, Schledzewski K, Goerdt S. GATA4-dependent organ-specific endothelial differentiation controls liver development and embryonic hematopoiesis. J Clin Invest 2017; 127:1099-1114. [PMID: 28218627 DOI: 10.1172/jci90086] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/20/2016] [Indexed: 12/23/2022] Open
Abstract
Microvascular endothelial cells (ECs) are increasingly recognized as organ-specific gatekeepers of their microenvironment. Microvascular ECs instruct neighboring cells in their organ-specific vascular niches through angiocrine factors, which include secreted growth factors (angiokines), extracellular matrix molecules, and transmembrane proteins. However, the molecular regulators that drive organ-specific microvascular transcriptional programs and thereby regulate angiodiversity are largely elusive. In contrast to other ECs, which form a continuous cell layer, liver sinusoidal ECs (LSECs) constitute discontinuous, permeable microvessels. Here, we have shown that the transcription factor GATA4 controls murine LSEC specification and function. LSEC-restricted deletion of Gata4 caused transformation of discontinuous liver sinusoids into continuous capillaries. Capillarization was characterized by ectopic basement membrane deposition, formation of a continuous EC layer, and increased expression of VE-cadherin. Correspondingly, ectopic expression of GATA4 in cultured continuous ECs mediated the downregulation of continuous EC-associated transcripts and upregulation of LSEC-associated genes. The switch from discontinuous LSECs to continuous ECs during embryogenesis caused liver hypoplasia, fibrosis, and impaired colonization by hematopoietic progenitor cells, resulting in anemia and embryonic lethality. Thus, GATA4 acts as master regulator of hepatic microvascular specification and acquisition of organ-specific vascular competence, which are indispensable for liver development. The data also establish an essential role of the hepatic microvasculature in embryonic hematopoiesis.
Collapse
|
182
|
Platelets prime hematopoietic and vascular niche to drive angiocrine-mediated liver regeneration. Signal Transduct Target Ther 2017; 2. [PMID: 29201496 PMCID: PMC5661617 DOI: 10.1038/sigtrans.2016.44] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In mammals, the livers regenerate after chemical injury or resection of hepatic lobe by hepatectomy. How liver regeneration is initiated after mass loss remains to be defined. Here, we report that following liver injury, activated platelets deploy SDF-1 and VEGF-A to stimulate CXCR7+ liver sinusoidal endothelial cell (LSEC) and VEGFR1+ myeloid cell, orchestrating hepatic regeneration. After carbon tetrachloride (CCl4) injection or hepatectomy, platelets and CD11b+VEGFR1+ myeloid cells were recruited LSEC, and liver regeneration in both models was impaired in thrombopoietin-deficient (Thpo-/-) mice lacking circulating platelets. This impeded regeneration phenotype was recapitulated in mice with either conditional ablation of Cxcr7 in LSEC (Cxcr7iΔ/iΔ) or Vegfr1 in myeloid cell (Vegfr1lysM/lysM). Both Vegfr1lysM/lysM and Cxcr7iΔ/iΔ mice exhibited suppressed expression of hepatocyte growth factor and Wnt2, two crucial trophogenic angiocrine factors instigating hepatocyte propagation. Of note, administration of recombinant thrombopoietin restored the prohibited liver regeneration in the tested genetic models. As such, our data suggest that platelets and myeloid cells jointly activate the vascular niche to produce pro-regenerative endothelial paracrine/angiocrine factors. Modulating this "hematopoietic-vascular niche" might help to develop regenerative therapy strategy for hepatic disorders.
Collapse
|
183
|
Wang S, Miller SR, Ober EA, Sadler KC. Making It New Again: Insight Into Liver Development, Regeneration, and Disease From Zebrafish Research. Curr Top Dev Biol 2017; 124:161-195. [PMID: 28335859 PMCID: PMC6450094 DOI: 10.1016/bs.ctdb.2016.11.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The adult liver of most vertebrates is predominantly comprised of hepatocytes. However, these cells must work in concert with biliary, stellate, vascular, and immune cells to accomplish the vast array of hepatic functions required for physiological homeostasis. Our understanding of liver development was accelerated as zebrafish emerged as an ideal vertebrate system to study embryogenesis. Through work in zebrafish and other models, it is now clear that the cells in the liver develop in a coordinated fashion during embryogenesis through a complex yet incompletely understood set of molecular guidelines. Zebrafish research has uncovered many key players that govern the acquisition of hepatic potential, cell fate, and plasticity. Although rare, some hepatobiliary diseases-especially biliary atresia-are caused by developmental defects; we discuss how research using zebrafish to study liver development has informed our understanding of and approaches to liver disease. The liver can be injured in response to an array of stressors including viral, mechanical/surgical, toxin-induced, immune-mediated, or inborn defects in metabolism. The liver has thus evolved the capacity to efficiently repair and regenerate. We discuss the emerging field of using zebrafish to study liver regeneration and highlight recent advances where zebrafish genetics and imaging approaches have provided novel insights into how cell plasticity contributes to liver regeneration.
Collapse
Affiliation(s)
- Shuang Wang
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Sophie R Miller
- Danish Stem Cell Center (DanStem), University of Copenhagen, Copenhagen N, Denmark
| | - Elke A Ober
- Danish Stem Cell Center (DanStem), University of Copenhagen, Copenhagen N, Denmark
| | - Kirsten C Sadler
- Icahn School of Medicine at Mount Sinai, New York, NY, United States; New York University Abu Dhabi, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
184
|
Hassanein W, Uluer MC, Langford J, Woodall JD, Cimeno A, Dhru U, Werdesheim A, Harrison J, Rivera-Pratt C, Klepfer S, Khalifeh A, Buckingham B, Brazio PS, Parsell D, Klassen C, Drachenberg C, Barth RN, LaMattina JC. Recellularization via the bile duct supports functional allogenic and xenogenic cell growth on a decellularized rat liver scaffold. Organogenesis 2016; 13:16-27. [PMID: 28029279 DOI: 10.1080/15476278.2016.1276146] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Recent years have seen a proliferation of methods leading to successful organ decellularization. In this experiment we examine the feasibility of a decellularized liver construct to support growth of functional multilineage cells. Bio-chamber systems were used to perfuse adult rat livers with 0.1% SDS for 24 hours yielding decellularized liver scaffolds. Initially, we recellularized liver scaffolds using a human tumor cell line (HepG2, introduced via the bile duct). Subsequent studies were performed using either human tumor cells co-cultured with human umbilical vein endothelial cells (HUVECs, introduced via the portal vein) or rat neonatal cell slurry (introduced via the bile duct). Bio-chambers were used to circulate oxygenated growth medium via the portal vein at 37C for 5-7 days. Human HepG2 cells grew readily on the scaffold (n = 20). HepG2 cells co-cultured with HUVECs demonstrated viable human endothelial lining with concurrent hepatocyte growth (n = 10). In the series of neonatal cell slurry infusion (n = 10), distinct foci of neonatal hepatocytes were observed to repopulate the parenchyma of the scaffold. The presence of cholangiocytes was verified by CK-7 positivity. Quantitative albumin measurement from the grafts showed increasing albumin levels after seven days of perfusion. Graft albumin production was higher than that observed in traditional cell culture. This data shows that rat liver scaffolds support human cell ingrowth. The scaffold likewise supported the engraftment and survival of neonatal rat liver cell slurry. Recellularization of liver scaffolds thus presents a promising model for functional liver engineering.
Collapse
Affiliation(s)
- Wessam Hassanein
- a University of Maryland School of Medicine, Department of Surgery , Division of Transplantation , Baltimore , MD , USA
| | - Mehmet C Uluer
- a University of Maryland School of Medicine, Department of Surgery , Division of Transplantation , Baltimore , MD , USA
| | - John Langford
- a University of Maryland School of Medicine, Department of Surgery , Division of Transplantation , Baltimore , MD , USA
| | - Jhade D Woodall
- a University of Maryland School of Medicine, Department of Surgery , Division of Transplantation , Baltimore , MD , USA
| | - Arielle Cimeno
- a University of Maryland School of Medicine, Department of Surgery , Division of Transplantation , Baltimore , MD , USA
| | - Urmil Dhru
- a University of Maryland School of Medicine, Department of Surgery , Division of Transplantation , Baltimore , MD , USA
| | - Avraham Werdesheim
- a University of Maryland School of Medicine, Department of Surgery , Division of Transplantation , Baltimore , MD , USA
| | - Joshua Harrison
- a University of Maryland School of Medicine, Department of Surgery , Division of Transplantation , Baltimore , MD , USA
| | - Carlos Rivera-Pratt
- a University of Maryland School of Medicine, Department of Surgery , Division of Transplantation , Baltimore , MD , USA
| | - Stephen Klepfer
- a University of Maryland School of Medicine, Department of Surgery , Division of Transplantation , Baltimore , MD , USA
| | - Ali Khalifeh
- a University of Maryland School of Medicine, Department of Surgery , Division of Transplantation , Baltimore , MD , USA
| | - Bryan Buckingham
- a University of Maryland School of Medicine, Department of Surgery , Division of Transplantation , Baltimore , MD , USA
| | - Philip S Brazio
- a University of Maryland School of Medicine, Department of Surgery , Division of Transplantation , Baltimore , MD , USA
| | - Dawn Parsell
- a University of Maryland School of Medicine, Department of Surgery , Division of Transplantation , Baltimore , MD , USA
| | - Charlie Klassen
- a University of Maryland School of Medicine, Department of Surgery , Division of Transplantation , Baltimore , MD , USA
| | - Cinthia Drachenberg
- b University of Maryland School of Medicine , Department of Pathology , Baltimore , MD , USA
| | - Rolf N Barth
- a University of Maryland School of Medicine, Department of Surgery , Division of Transplantation , Baltimore , MD , USA
| | - John C LaMattina
- a University of Maryland School of Medicine, Department of Surgery , Division of Transplantation , Baltimore , MD , USA
| |
Collapse
|
185
|
Ding BS, Liu CH, Sun Y, Chen Y, Swendeman SL, Jung B, Chavez D, Cao Z, Christoffersen C, Nielsen LB, Schwab SR, Rafii S, Hla T. HDL activation of endothelial sphingosine-1-phosphate receptor-1 (S1P 1) promotes regeneration and suppresses fibrosis in the liver. JCI Insight 2016; 1:e87058. [PMID: 28018969 PMCID: PMC5161208 DOI: 10.1172/jci.insight.87058] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Regeneration of hepatic sinusoidal vasculature is essential for non-fibrotic liver regrowth and restoration of its metabolic capacity. However, little is known about how this specialized vascular niche is regenerated. Here we show that activation of endothelial sphingosine-1-phosphate receptor-1 (S1P1) by its natural ligand bound to HDL (HDL-S1P) induces liver regeneration and curtails fibrosis. In mice lacking HDL-S1P, liver regeneration after partial hepatectomy was impeded and associated with aberrant vascular remodeling, thrombosis and peri-sinusoidal fibrosis. Notably, this "maladaptive repair" phenotype was recapitulated in mice that lack S1P1 in the endothelium. Reciprocally, enhanced plasma levels of HDL-S1P or administration of SEW2871, a pharmacological agonist specific for S1P1 enhanced regeneration of metabolically functional vasculature and alleviated fibrosis in mouse chronic injury and cholestasis models. This study shows that natural and pharmacological ligands modulate endothelial S1P1 to stimulate liver regeneration and inhibit fibrosis, suggesting that activation of this pathway may be a novel therapeutic strategy for liver fibrosis.
Collapse
Affiliation(s)
- Bi-Sen Ding
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.,Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Catherine H Liu
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, New York, USA
| | - Yue Sun
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yutian Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Steven L Swendeman
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, New York, USA.,Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Bongnam Jung
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Deebly Chavez
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Zhongwei Cao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.,Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark.,Department of Biomedical Sciences
| | - Lars Bo Nielsen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark.,Department of Biomedical Sciences.,Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Susan R Schwab
- Department of Pathology, Skirball Institute, New York University School of Medicine, New York, New York, USA
| | - Shahin Rafii
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Timothy Hla
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, New York, USA.,Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
186
|
Abstract
Defined genetic models based on human pluripotent stem cells have opened new avenues for understanding disease mechanisms and drug screening. Many of these models assume cell-autonomous mechanisms of disease but it is possible that disease phenotypes or drug responses will only be evident if all cellular and extracellular components of a tissue are present and functionally mature. To derive optimal benefit from such models, complex multicellular structures with vascular components that mimic tissue niches will thus likely be necessary. Here we consider emerging research creating human tissue mimics and provide some recommendations for moving the field forward.
Collapse
|
187
|
Gérard C, Tys J, Lemaigre FP. Gene regulatory networks in differentiation and direct reprogramming of hepatic cells. Semin Cell Dev Biol 2016; 66:43-50. [PMID: 27979774 DOI: 10.1016/j.semcdb.2016.12.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/07/2016] [Indexed: 12/14/2022]
Abstract
Liver development proceeds by sequential steps during which gene regulatory networks (GRNs) determine differentiation and maturation of hepatic cells. Characterizing the architecture and dynamics of these networks is essential for understanding how cell fate decisions are made during development, and for recapitulating these processes during in vitro production of liver cells for toxicology studies, disease modelling and regenerative therapy. Here we review the GRNs that control key steps of liver development and lead to differentiation of hepatocytes and cholangiocytes in mammals. We focus on GRNs determining cell fate decisions and analyse subcircuitry motifs that may confer specific dynamic properties to the networks. Finally, we put our analysis in the perspective of recent attempts to directly reprogram cells to hepatocytes by forced expression of transcription factors.
Collapse
Affiliation(s)
- Claude Gérard
- Université catholique de Louvain, de Duve Institute, Avenue Hippocrate 75, 1200 Brussels, Belgium.
| | - Janne Tys
- Université catholique de Louvain, de Duve Institute, Avenue Hippocrate 75, 1200 Brussels, Belgium.
| | - Frédéric P Lemaigre
- Université catholique de Louvain, de Duve Institute, Avenue Hippocrate 75, 1200 Brussels, Belgium.
| |
Collapse
|
188
|
Talavera-Adame D, Woolcott OO, Ignatius-Irudayam J, Arumugaswami V, Geller DH, Dafoe DC. Effective endothelial cell and human pluripotent stem cell interactions generate functional insulin-producing beta cells. Diabetologia 2016; 59:2378-2386. [PMID: 27567623 PMCID: PMC5506104 DOI: 10.1007/s00125-016-4078-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/01/2016] [Indexed: 12/27/2022]
Abstract
AIMS/HYPOTHESIS Endothelial cells (ECs) play an essential role in pancreatic organogenesis. We hypothesise that effective in vitro interactions between human microvascular endothelial cells (HMECs) and human pluripotent stem cells (hPSCs) results in the generation of functional pancreatic beta cells. METHODS Embryoid bodies (EBs) derived from hPSCs were cultured alone (controls) or with ECs in collagen gels. Subsequently, cells were analysed for pancreatic beta cell markers, and then isolated and expanded. Insulin secretion in response to glucose was evaluated in vitro by static and dynamic (perifusion) assays, and in vivo by EB transplantation into immunodeficient mice. RESULTS Co-cultured EBs had a higher expression of mature beta cells markers and enhanced insulin secretion in vitro, compared with controls. In mice, transplanted EBs had higher levels of human C-peptide secretion with a significant reduction in hyperglycaemia after the selective destruction of native pancreatic beta cells. In addition, there was significant in vitro upregulation of bone morphogenetic proteins 2 and 4 (BMP-2, 4) in co-cultured cells, compared with controls. CONCLUSIONS/INTERPRETATION ECs provide essential signalling in vitro, such as activation of the BMP pathway, for derivation of functional insulin-producing beta cells from hPSCs.
Collapse
Affiliation(s)
- Dodanim Talavera-Adame
- Comprehensive Transplant Center, Department of Surgery, Cedars-Sinai Medical Center, 8900 Beverly Boulevard, 251E, Los Angeles, CA, 90048, USA.
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Orison O Woolcott
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Joseph Ignatius-Irudayam
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Vaithilingaraja Arumugaswami
- Comprehensive Transplant Center, Department of Surgery, Cedars-Sinai Medical Center, 8900 Beverly Boulevard, 251E, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - David H Geller
- Pediatric Endocrinology, Children's Hospital, Los Angeles, CA, USA
| | - Donald C Dafoe
- Comprehensive Transplant Center, Department of Surgery, Cedars-Sinai Medical Center, 8900 Beverly Boulevard, 251E, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
189
|
DeSesso JM. Vascular ontogeny within selected thoracoabdominal organs and the limbs. Reprod Toxicol 2016; 70:3-20. [PMID: 27810254 DOI: 10.1016/j.reprotox.2016.10.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/21/2016] [Accepted: 10/28/2016] [Indexed: 01/03/2023]
Abstract
The cardiovascular system is fundamental to life. Its vessels are the conduits for delivery of nutrients and oxygen to organs and the removal of wastes. During embryonic development, the vascular system is instrumental in the formation of organs. It contributes to the form and pattern of organs as diverse as the limbs and the gonads. Recent advances in molecular biology and genomics have afforded great insight to the control of vascular development at subcellular levels of organization. Nevertheless, there is little assembled information concerning the vascular development of the organ systems of the body. This paper begins by reviewing the modes of formation of embryonic blood vessels. This is followed by summaries of the ontogeny of the vasculature that supplies selected major thoracic and abdominal organs (heart, gut, liver, gonads, and kidney). The paper concludes with a description of the arterial development of the upper and lower extremities.
Collapse
Affiliation(s)
- John M DeSesso
- Exponent, Inc., Alexandria, VA, United States; Georgetown University School of Medicine Washington, District of Columbia, United States.
| |
Collapse
|
190
|
Cañete A, Comaills V, Prados I, Castro AM, Hammad S, Ybot-Gonzalez P, Bockamp E, Hengstler JG, Gottgens B, Sánchez MJ. Characterization of a Fetal Liver Cell Population Endowed with Long-Term Multiorgan Endothelial Reconstitution Potential. Stem Cells 2016; 35:507-521. [PMID: 27615355 PMCID: PMC5298023 DOI: 10.1002/stem.2494] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 07/27/2016] [Accepted: 08/10/2016] [Indexed: 12/26/2022]
Abstract
Stable reconstitution of vascular endothelial beds upon transplantation of progenitor cells represents an important challenge due to the paucity and generally limited integration/expansion potential of most identified vascular related cell subsets. We previously showed that mouse fetal liver (FL) hemato/vascular cells from day 12 of gestation (E12), expressing the Stem Cell Leukaemia (SCL) gene enhancer transgene (SCL‐PLAP+ cells), had robust endothelial engraftment potential when transferred to the blood stream of newborns or adult conditioned recipients, compared to the scarce vascular contribution of adult bone marrow cells. However, the specific SCL‐PLAP+ hematopoietic or endothelial cell subset responsible for the long‐term reconstituting endothelial cell (LTR‐EC) activity and its confinement to FL developmental stages remained unknown. Using a busulfan‐treated newborn transplantation model, we show that LTR‐EC activity is restricted to the SCL‐PLAP+VE‐cadherin+CD45− cell population, devoid of hematopoietic reconstitution activity and largely composed by Lyve1+ endothelial‐committed cells. SCL‐PLAP+ Ve‐cadherin+CD45− cells contributed to the liver sinusoidal endothelium and also to the heart, kidney and lung microvasculature. LTR‐EC activity was detected at different stages of FL development, yet marginal activity was identified in the adult liver, revealing unknown functional differences between fetal and adult liver endothelial/endothelial progenitors. Importantly, the observations that expanding donor‐derived vascular grafts colocalize with proliferating hepatocyte‐like cells and participate in the systemic circulation, support their functional integration into young livers. These findings offer new insights into the engraftment, phonotypical, and developmental characterization of a novel endothelial/endothelial progenitor cell subtype with multiorgan LTR‐EC activity, potentially instrumental for the treatment/genetic correction of vascular diseases. Stem Cells2017;35:507–521
Collapse
Affiliation(s)
- Ana Cañete
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas (CSIC), Junta de Andalucía (JA), Universidad Pablo de Olavide (UPO), Sevilla, Spain
| | - Valentine Comaills
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas (CSIC), Junta de Andalucía (JA), Universidad Pablo de Olavide (UPO), Sevilla, Spain
| | - Isabel Prados
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas (CSIC), Junta de Andalucía (JA), Universidad Pablo de Olavide (UPO), Sevilla, Spain
| | - Ana María Castro
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas (CSIC), Junta de Andalucía (JA), Universidad Pablo de Olavide (UPO), Sevilla, Spain
| | - Seddik Hammad
- Faculty of Veterinary Medicine, Department of Forensic Medicine and Veterinary Toxicology, South Valley University, Qena, Egypt.,Leibniz Research Center for Working Environment and Human Factors (IfADo), TU Dortmund University, Dortmund, Germany
| | - Patricia Ybot-Gonzalez
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain
| | - Ernesto Bockamp
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Jan G Hengstler
- Leibniz Research Center for Working Environment and Human Factors (IfADo), TU Dortmund University, Dortmund, Germany
| | - Bertie Gottgens
- Cambridge Institute for Medical Research & Wellcome Trust and MRC Cambridge Stem Cell Institute, Cambridge University, United Kingdom
| | - María José Sánchez
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas (CSIC), Junta de Andalucía (JA), Universidad Pablo de Olavide (UPO), Sevilla, Spain
| |
Collapse
|
191
|
James A, Lee C, Williams AM, Angileri K, Lathrop KL, Gross JM. The hyaloid vasculature facilitates basement membrane breakdown during choroid fissure closure in the zebrafish eye. Dev Biol 2016; 419:262-272. [PMID: 27634568 DOI: 10.1016/j.ydbio.2016.09.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 09/09/2016] [Accepted: 09/09/2016] [Indexed: 12/22/2022]
Abstract
A critical aspect of vertebrate eye development is closure of the choroid fissure (CF). Defects in CF closure result in colobomas, which are a significant cause of childhood blindness worldwide. Despite the growing number of mutated loci associated with colobomas, we have a limited understanding of the cell biological underpinnings of CF closure. Here, we utilize the zebrafish embryo to identify key phases of CF closure and regulators of the process. Utilizing Laminin-111 as a marker for the basement membrane (BM) lining the CF, we determine the spatial and temporal patterns of BM breakdown in the CF, a prerequisite for CF closure. Similarly, utilizing a combination of in vivo time-lapse imaging, β-catenin immunohistochemistry and F-actin staining, we determine that tissue fusion, which serves to close the fissure, follows BM breakdown closely. Periocular mesenchyme (POM)-derived endothelial cells, which migrate through the CF to give rise to the hyaloid vasculature, possess distinct actin foci that correlate with regions of BM breakdown. Disruption of talin1, which encodes a regulator of the actin cytoskeleton, results in colobomas and these correlate with structural defects in the hyaloid vasculature and defects in BM breakdown. cloche mutants, which entirely lack a hyaloid vasculature, also possess defects in BM breakdown in the CF. Taken together, these data support a model in which the hyaloid vasculature and/or the POM-derived endothelial cells that give rise to the hyaloid vasculature contribute to BM breakdown during CF closure.
Collapse
Affiliation(s)
- Andrea James
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin TX, 78712
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Chanjae Lee
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin TX, 78712
| | - Andre M Williams
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin TX, 78712
| | - Krista Angileri
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin TX, 78712
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Kira L Lathrop
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15213
| | - Jeffrey M Gross
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin TX, 78712
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
- Department of Developmental Biology, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| |
Collapse
|
192
|
Wan AC. Recapitulating Cell–Cell Interactions for Organoid Construction – Are Biomaterials Dispensable? Trends Biotechnol 2016; 34:711-721. [DOI: 10.1016/j.tibtech.2016.02.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 02/23/2016] [Accepted: 02/26/2016] [Indexed: 12/27/2022]
|
193
|
Ramasamy SK, Kusumbe AP, Itkin T, Gur-Cohen S, Lapidot T, Adams RH. Regulation of Hematopoiesis and Osteogenesis by Blood Vessel-Derived Signals. Annu Rev Cell Dev Biol 2016; 32:649-675. [PMID: 27576121 DOI: 10.1146/annurev-cellbio-111315-124936] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In addition to their conventional role as a versatile transport system, blood vessels provide signals controlling organ development, regeneration, and stem cell behavior. In the skeletal system, certain capillaries support perivascular osteoprogenitor cells and thereby control bone formation. Blood vessels are also a critical component of niche microenvironments for hematopoietic stem cells. Here we discuss key pathways and factors controlling endothelial cell behavior in bone, the role of vessels in osteogenesis, and the nature of vascular stem cell niches in bone marrow.
Collapse
Affiliation(s)
- Saravana K Ramasamy
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48169 Münster, Germany; .,Faculty of Medicine, University of Münster, D-48149 Münster, Germany
| | - Anjali P Kusumbe
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48169 Münster, Germany; .,Faculty of Medicine, University of Münster, D-48149 Münster, Germany
| | - Tomer Itkin
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel;
| | - Shiri Gur-Cohen
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel;
| | - Tsvee Lapidot
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel;
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48169 Münster, Germany; .,Faculty of Medicine, University of Münster, D-48149 Münster, Germany
| |
Collapse
|
194
|
The endoderm indirectly influences morphogenetic movements of the zebrafish head kidney through the posterior cardinal vein and VegfC. Sci Rep 2016; 6:30677. [PMID: 27477767 PMCID: PMC4967926 DOI: 10.1038/srep30677] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/07/2016] [Indexed: 02/01/2023] Open
Abstract
Integration of blood vessels and organ primordia determines organ shape and function. The head kidney in the zebrafish interacts with the dorsal aorta (DA) and the posterior cardinal vein (PCV) to achieve glomerular filtration and definitive hematopoiesis, respectively. How the head kidney co-develops with both the axial artery and vein remains unclear. We found that in endodermless sox32-deficient embryos, the head kidney associated with the PCV but not the DA. Disrupted convergent migration of the PCV and the head kidney in sox32-deficient embryos was rescued in a highly coordinated fashion through the restoration of endodermal cells. Moreover, grafted endodermal cells abutted the host PCV endothelium in the transplantation assay. Interestingly, the severely-disrupted head kidney convergence in the sox32-deficient embryo was suppressed by both the cloche mutation and the knockdown of endothelial genes, indicating that an interaction between the endoderm and the PCV restricts the migration of the head kidney. Furthermore, knockdown of either vegfC or its receptor vegfr3 suppressed the head kidney convergence defect in endodermless embryos and perturbed the head kidney-PCV association in wild-type embryos. Our findings thus underscore a role for PCV and VegfC in patterning the head kidney prior to organ assembly and function.
Collapse
|
195
|
Rafii S, Ginsberg M, Scandura J, Butler JM, Ding BS. Transplantation of Endothelial Cells to Mitigate Acute and Chronic Radiation Injury to Vital Organs. Radiat Res 2016; 186:196-202. [PMID: 27459700 DOI: 10.1667/rr14461.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Current therapeutic approaches for treatment of exposure to radiation involve the use of antioxidants, chelating agents, recombinant growth factors and transplantation of stem cells (e.g., hematopoietic stem cell transplantation). However, exposure to high-dose radiation is associated with severe damage to the vasculature of vital organs, often leading to impaired healing, tissue necrosis, thrombosis and defective regeneration caused by aberrant fibrosis. It is very unlikely that infusion of protective chemicals will reverse severe damage to the vascular endothelial cells (ECs). The role of irradiated vasculature in mediating acute and chronic radiation syndromes has not been fully appreciated or well studied. New approaches are necessary to replace and reconstitute ECs in organs that are irreversibly damaged by radiation. We have set forth the novel concept that ECs provide paracrine signals, also known as angiocrine signals, which not only promote healing of irradiated tissue but also direct organ regeneration without provoking fibrosis. We have developed innovative technologies that enable manufacturing and banking of human GMP-grade ECs. These ECs can be transplanted intravenously to home to and engraft to injured tissues where they augment organ repair, while preventing maladaptive fibrosis. In the past, therapeutic transplantation of ECs was not possible due to a shortage of availability of suitable donor cell sources and preclinical models, a lack of understanding of the immune privilege of ECs, and inadequate methodologies for expansion and banking of engraftable ECs. Recent advances made by our group as well as other laboratories have breached the most significant of these obstacles with the development of technologies to manufacture clinical-scale quantities of GMP-grade and human ECs in culture, including genetically diverse reprogrammed human amniotic cells into vascular ECs (rAC-VECs) or human pluripotent stem cells into vascular ECs (iVECs). This approach provides a path to therapeutic EC transplantation that can be infused concomitantly or sequentially with hematopoietic stem cell transplantation more than 24 h after irradiation to support multi-organ regeneration, thereby improving immediate and long-term survival, while limiting long-term morbidity resulting from nonregenerative damage repair pathways.
Collapse
Affiliation(s)
- Shahin Rafii
- Weill Cornell Medical College Ansary Stem Cell Institute, Department of Medicine, Division of Regenerative Medicine, New York, New York
| | - Michael Ginsberg
- Weill Cornell Medical College Ansary Stem Cell Institute, Department of Medicine, Division of Regenerative Medicine, New York, New York
| | - Joseph Scandura
- Weill Cornell Medical College Ansary Stem Cell Institute, Department of Medicine, Division of Regenerative Medicine, New York, New York
| | - Jason M Butler
- Weill Cornell Medical College Ansary Stem Cell Institute, Department of Medicine, Division of Regenerative Medicine, New York, New York
| | - Bi-Sen Ding
- Weill Cornell Medical College Ansary Stem Cell Institute, Department of Medicine, Division of Regenerative Medicine, New York, New York
| |
Collapse
|
196
|
Shinozawa T, Yoshikawa HY, Takebe T. Reverse engineering liver buds through self-driven condensation and organization towards medical application. Dev Biol 2016; 420:221-229. [PMID: 27364470 DOI: 10.1016/j.ydbio.2016.06.036] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 05/24/2016] [Accepted: 06/25/2016] [Indexed: 12/15/2022]
Abstract
The self-organizing tissue-based approach coupled with induced pluripotent stem (iPS) cell technology is evolving as a promising field for designing organoids in culture and is expected to achieve valuable practical outcomes in regenerative medicine and drug development. Organoids show properties of functional organs and represent an alternative to cell models in conventional two-dimensional differentiation platforms; moreover, organoids can be used to investigate mechanisms of development and disease, drug discovery and toxicity assessment. Towards a more complex and advanced organoid model, it is essential to incorporate multiple cell lineages including developing vessels. Using a self-condensation method, we recently demonstrated self-organizing "organ buds" of diverse systems together with human mesenchymal and endothelial progenitors, proposing a new reverse engineering method to generate a more complex organoid structure. In this section, we review characters of organ bud technology based on two important principles: self-condensation and self-organization focusing on liver bud as an example, and discuss their practicality in regenerative medicine and potential as research tools for developmental biology and drug discovery.
Collapse
Affiliation(s)
- Tadahiro Shinozawa
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Hiroshi Y Yoshikawa
- Department of Chemistry, Saitama University, Shimo-okubo 255, Sakura-ku, Saitama 338-8570, Japan.
| | - Takanori Takebe
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan; PRESTO, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan.
| |
Collapse
|
197
|
Hindley CJ, Cordero-Espinoza L, Huch M. Organoids from adult liver and pancreas: Stem cell biology and biomedical utility. Dev Biol 2016; 420:251-261. [PMID: 27364469 DOI: 10.1016/j.ydbio.2016.06.039] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 06/26/2016] [Accepted: 06/26/2016] [Indexed: 01/02/2023]
Abstract
The liver and pancreas are critical organs maintaining whole body metabolism. Historically, the expansion of adult-derived cells from these organs in vitro has proven challenging and this in turn has hampered studies of liver and pancreas stem cell biology, as well as being a roadblock to disease modelling and cell replacement therapies for pathologies in these organs. Recently, defined culture conditions have been described which allow the in vitro culture and manipulation of adult-derived liver and pancreatic material. Here we review these systems and assess their physiological relevance, as well as their potential utility in biomedicine.
Collapse
Affiliation(s)
- Christopher J Hindley
- Wellcome Trust/Cancer Research UK Gurdon Institute, Henry Wellcome Building of Cancer and Developmental Biology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; The Cavendish Laboratory, Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, UK
| | - Lucía Cordero-Espinoza
- Wellcome Trust/Cancer Research UK Gurdon Institute, Henry Wellcome Building of Cancer and Developmental Biology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; Wellcome Trust/Medical Research Council Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Meritxell Huch
- Wellcome Trust/Cancer Research UK Gurdon Institute, Henry Wellcome Building of Cancer and Developmental Biology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; Wellcome Trust/Medical Research Council Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK.
| |
Collapse
|
198
|
Azizoglu DB, Cleaver O. Blood vessel crosstalk during organogenesis-focus on pancreas and endothelial cells. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2016; 5:598-617. [PMID: 27328421 DOI: 10.1002/wdev.240] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/23/2016] [Accepted: 04/16/2016] [Indexed: 01/02/2023]
Abstract
Blood vessels form a highly branched, interconnected, and largely stereotyped network of tubes that sustains every organ and tissue in vertebrates. How vessels come to take on their particular architecture, or how they are 'patterned,' and in turn, how they influence surrounding tissues are fundamental questions of organogenesis. Decades of work have begun to elucidate how endothelial progenitors arise and home to precise locations within tissues, integrating attractive and repulsive cues to build vessels where they are needed. Conversely, more recent findings have revealed an exciting facet of blood vessel interaction with tissues, where vascular cells provide signals to developing organs and progenitors therein. Here, we discuss the exchange of reciprocal signals between endothelial cells and neighboring tissues during embryogenesis, with a special focus on the developing pancreas. Understanding the mechanisms driving both sides of these interactions will be crucial to the development of therapies, from improving organ regeneration to efficient production of cell based therapies. Specifically, elucidating the interface of the vasculature with pancreatic lineages, including endocrine cells, will instruct approaches such as generation of replacement beta cells for Type I diabetes. WIREs Dev Biol 2016, 5:598-617. doi: 10.1002/wdev.240 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- D Berfin Azizoglu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
199
|
Touboul T, Chen S, To CC, Mora-Castilla S, Sabatini K, Tukey RH, Laurent LC. Stage-specific regulation of the WNT/β-catenin pathway enhances differentiation of hESCs into hepatocytes. J Hepatol 2016; 64:1315-26. [PMID: 26921690 PMCID: PMC5010388 DOI: 10.1016/j.jhep.2016.02.028] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 02/02/2016] [Accepted: 02/03/2016] [Indexed: 01/25/2023]
Abstract
BACKGROUND & AIMS Hepatocytes differentiated from human embryonic stem cells (hESCs) have the potential to overcome the shortage of primary hepatocytes for clinical use and drug development. Many strategies for this process have been reported, but the functionality of the resulting cells is incomplete. We hypothesize that the functionality of hPSC-derived hepatocytes might be improved by making the differentiation method more similar to normal in vivo hepatic development. METHODS We tested combinations of growth factors and small molecules targeting candidate signaling pathways culled from the literature to identify optimal conditions for differentiation of hESCs to hepatocytes, using qRT-PCR for stage-specific markers to identify the best conditions. Immunocytochemistry was then used to validate the selected conditions. Finally, induction of expression of metabolic enzymes in terminally differentiated cells was used to assess the functionality of the hESC-derived hepatocytes. RESULTS Optimal differentiation of hESCs was attained using a 5-stage protocol. After initial induction of definitive endoderm (stage 1), we showed that inhibition of the WNT/β-catenin pathway during the 2nd and 3rd stages of differentiation was required to specify first posterior foregut, and then hepatic gut cells. In contrast, during the 4th stage of differentiation, we found that activation of the WNT/β-catenin pathway allowed generation of proliferative bipotent hepatoblasts, which then were efficiently differentiated into hepatocytes in the 5th stage by dual inhibition of TGF-β and NOTCH signaling. CONCLUSION Here, we show that stage-specific regulation of the WNT/β-catenin pathway results in improved differentiation of hESCs to functional hepatocytes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Louise C Laurent
- Department of Reproductive Medicine, UC San Diego, La Jolla, CA 92037, United States.
| |
Collapse
|
200
|
Rafii S, Butler JM, Ding BS. Angiocrine functions of organ-specific endothelial cells. Nature 2016; 529:316-25. [PMID: 26791722 DOI: 10.1038/nature17040] [Citation(s) in RCA: 683] [Impact Index Per Article: 75.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 11/26/2015] [Indexed: 12/13/2022]
Abstract
Endothelial cells that line capillaries are not just passive conduits for delivering blood. Tissue-specific endothelium establishes specialized vascular niches that deploy sets of growth factors, known as angiocrine factors. These cues participate actively in the induction, specification, patterning and guidance of organ regeneration, as well as in the maintainance of homeostasis and metabolism. When upregulated following injury, they orchestrate self-renewal and differentiation of tissue-specific resident stem and progenitor cells into functional organs. Uncovering the mechanisms by which organotypic endothelium distributes physiological levels of angiocrine factors both spatially and temporally will lay the foundation for clinical trials that promote organ repair without scarring.
Collapse
Affiliation(s)
- Shahin Rafii
- Ansary Stem Cell Institute, Department of Medicine, Division of Regenerative Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, New York 10065, USA
| | - Jason M Butler
- Ansary Stem Cell Institute, Department of Medicine, Division of Regenerative Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, New York 10065, USA
| | - Bi-Sen Ding
- Ansary Stem Cell Institute, Department of Medicine, Division of Regenerative Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, New York 10065, USA
| |
Collapse
|