101
|
Aiyelabegan HT, Zaidi SSZ, Fanuel S, Eatemadi A, Ebadi MTK, Sadroddiny E. Albumin-based biomaterial for lung tissue engineering applications. INT J POLYM MATER PO 2016. [DOI: 10.1080/00914037.2016.1180610] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
102
|
A panel data set on harvest and perfusion decellularization of porcine rectus abdominis. Data Brief 2016; 7:1375-82. [PMID: 27158653 PMCID: PMC4845073 DOI: 10.1016/j.dib.2016.04.018] [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: 03/01/2016] [Revised: 04/02/2016] [Accepted: 04/06/2016] [Indexed: 11/26/2022] Open
Abstract
In this dataset, we particularly depicted the harvest and perfusion decellularization of porcine rectus abdominis (RA), accompanied with displaying of the retained vascular trees within the perfusion-decellularized skeletal muscle matrix (pM-ECM) using vascular corrosion casting. In addition, several important tips for successful pM-ECM preparation were emphasized, which including using anatomically isolated skeletal muscle as tissue source with all main feeding and draining vessels perfused, preserving the internal microcirculation availability, aseptic technique and pyrogen free in all steps, sequential perfusion via artery or vein, and longtime washing after decellularization. The data are supplemental to our original research article describing detailed associations of pM-ECM as a clinically relevant scale, three-dimensional scaffold with a vascular network template for tissue-specific regeneration, “Perfusion-decellularized skeletal muscle as a three-dimensional scaffold with a vascular network template” Zhang et al. (2016) [1].
Collapse
|
103
|
Attanasio C, Latancia MT, Otterbein LE, Netti PA. Update on Renal Replacement Therapy: Implantable Artificial Devices and Bioengineered Organs. TISSUE ENGINEERING PART B-REVIEWS 2016; 22:330-40. [PMID: 26905099 DOI: 10.1089/ten.teb.2015.0467] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Recent advances in the fields of artificial organs and regenerative medicine are now joining forces in the areas of organ transplantation and bioengineering to solve continued challenges for patients with end-stage renal disease. The waiting lists for those needing a transplant continue to exceed demand. Dialysis, while effective, brings different challenges, including quality of life and susceptibility to infection. Unfortunately, the majority of research outputs are far from delivering satisfactory solutions. Current efforts are focused on providing a self-standing device able to recapitulate kidney function. In this review, we focus on two remarkable innovations that may offer significant clinical impact in the field of renal replacement therapy: the implantable artificial renal assist device (RAD) and the transplantable bioengineered kidney. The artificial RAD strategy utilizes micromachining techniques to fabricate a biohybrid system able to mimic renal morphology and function. The current trend in kidney bioengineering exploits the structure of the native organ to produce a kidney that is ready to be transplanted. Although these two systems stem from different technological approaches, they are both designed to be implantable, long lasting, and free standing to allow patients with kidney failure to be autonomous. However, for both of them, there are relevant issues that must be addressed before translation into clinical use and these are discussed in this review.
Collapse
Affiliation(s)
- Chiara Attanasio
- 1 Center for Advanced Biomaterials for Health Care, IIT@CRIB, Istituto Italiano di Tecnologia , Napoli, Italy
| | - Marcela T Latancia
- 2 Department of Surgery, Transplant Institute , Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Leo E Otterbein
- 2 Department of Surgery, Transplant Institute , Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Paolo A Netti
- 1 Center for Advanced Biomaterials for Health Care, IIT@CRIB, Istituto Italiano di Tecnologia , Napoli, Italy
| |
Collapse
|
104
|
Poornejad N, Schaumann LB, Buckmiller EM, Roeder BL, Cook AD. Current Cell-Based Strategies for Whole Kidney Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2016; 22:358-370. [PMID: 26905375 DOI: 10.1089/ten.teb.2015.0520] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chronic kidney diseases affect thousands of people worldwide. Although hemodialysis alleviates the situation by filtering the patient's blood, it does not replace other kidney functions such as hormone release or homeostasis regulation. Consequently, orthotopic transplantation of donor organs is the ultimate treatment for patients suffering from end-stage renal failure. Unfortunately, the number of patients on the waiting list far exceeds the number of donors. In addition, recipients must remain on immunosuppressive medications for the remainder of their lives, which increases the risk of morbidity due to their weakened immune system. Despite recent advancements in whole organ transplantation, 40% of recipients will face rejection of implanted organs with a life expectancy of only 10 years. Bioengineered patient-specific kidneys could be an inexhaustible source of healthy kidneys without the risk of immune rejection. The purpose of this article is to review the pros and cons of several bioengineering strategies used in recent years and their unresolved issues. These strategies include repopulation of natural scaffolds with a patient's cells, de-novo generation of kidneys using patient-induced pluripotent stem cells combined with stepwise differentiation, and the creation of a patient's kidney in the embryos of other mammalian species.
Collapse
Affiliation(s)
- Nafiseh Poornejad
- 1 Department of Chemical Engineering, Brigham Young University , Provo, Utah
| | - Lara B Schaumann
- 1 Department of Chemical Engineering, Brigham Young University , Provo, Utah
| | - Evan M Buckmiller
- 2 Department of Genetics and Biotechnology, Brigham Young University , Provo, Utah
| | | | - Alonzo D Cook
- 1 Department of Chemical Engineering, Brigham Young University , Provo, Utah
| |
Collapse
|
105
|
Montserrat N, Garreta E, Izpisua Belmonte JC. Regenerative strategies for kidney engineering. FEBS J 2016; 283:3303-24. [DOI: 10.1111/febs.13704] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 01/22/2016] [Accepted: 03/01/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Nuria Montserrat
- Pluripotent Stem Cells and Activation of Endogenous Tissue Programs for Organ Regeneration (PR Lab) Institute for Bioengineering of Catalonia (IBEC) Barcelona Spain
- Networking Biomedical Research Center in Bioengineering Biomaterials and Nanomedicine (CIBER‐BBN) Madrid Spain
| | - Elena Garreta
- Pluripotent Stem Cells and Activation of Endogenous Tissue Programs for Organ Regeneration (PR Lab) Institute for Bioengineering of Catalonia (IBEC) Barcelona Spain
| | | |
Collapse
|
106
|
McKee RA, Wingert RA. Repopulating Decellularized Kidney Scaffolds: An Avenue for Ex Vivo Organ Generation. MATERIALS 2016; 9. [PMID: 27375844 PMCID: PMC4927010 DOI: 10.3390/ma9030190] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Recent research has shown that fully developed organs can be decellularized, resulting in a complex scaffold and extracellular matrix (ECM) network capable of being populated with other cells. This work has resulted in a growing field in bioengineering focused on the isolation, characterization, and modification of organ derived acellular scaffolds and their potential to sustain and interact with new cell populations, a process termed reseeding. In this review, we cover contemporary advancements in the bioengineering of kidney scaffolds including novel work showing that reseeded donor scaffolds can be transplanted and can function in recipients using animal models. Several major areas of the field are taken into consideration, including the decellularization process, characterization of acellular and reseeded scaffolds, culture conditions, and cell sources. Finally, we discuss future avenues based on the advent of 3D bioprinting and recent developments in kidney organoid cultures as well as animal models of renal genesis. The ongoing mergers and collaborations between these fields hold the potential to produce functional kidneys that can be generated ex vivo and utilized for kidney transplantations in patients suffering with renal disease.
Collapse
|
107
|
Poornejad N, Momtahan N, Salehi ASM, Scott DR, Fronk CA, Roeder BL, Reynolds PR, Bundy BC, Cook AD. Efficient decellularization of whole porcine kidneys improves reseeded cell behavior. Biomed Mater 2016; 11:025003. [DOI: 10.1088/1748-6041/11/2/025003] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
108
|
Zhang J, Hu ZQ, Turner NJ, Teng SF, Cheng WY, Zhou HY, Zhang L, Hu HW, Wang Q, Badylak SF. Perfusion-decellularized skeletal muscle as a three-dimensional scaffold with a vascular network template. Biomaterials 2016; 89:114-26. [PMID: 26963901 DOI: 10.1016/j.biomaterials.2016.02.040] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/07/2016] [Accepted: 02/23/2016] [Indexed: 11/25/2022]
Abstract
There exists a great need for repair grafts with similar volume to human skeletal muscle that can promote the innate ability of muscle to regenerate following volumetric muscle loss. Perfusion decellularization is an attractive technique for extracellular matrix (ECM) scaffold from intact mammalian organ or tissue which has been successfully used in tissue reconstruction. The perfusion-decellularization of skeletal muscle has been poorly assessed and characterized, but the bioactivity and functional capacity of the obtained perfusion skeletal muscle ECM (pM-ECM) to remodel in vivo is unknown. In the present study, pM-ECM was prepared from porcine rectus abdominis (RA). Perfusion-decellularization of porcine RA effectively removed cellular and nuclear material while retaining the intricate three-dimensional microarchitecture and vasculature networks of the native RA, and many of the bioactive ECM components and mechanical properties. In vivo, partial-thickness abdominal wall defects in rats repaired with pM-ECM showed improved neovascularization, myogenesis and functional recellularization compared to porcine-derived small intestinal submucosa (SIS). These findings show the biologic potential of RA pM-ECM as a scaffold for supporting site appropriate, tissue reconstruction, and provide a better understanding of the importance maintaining the tissue-specific complex three-dimensional architecture of ECM during decellularization and regeneration.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Surgery, Shanghai Chang Zheng Hospital, Second Military Medical University, Shanghai 200003, PR China; Department of Regenerative Medicine, Shanghai Zhabei District Central Hospital, Shanghai 200072, PR China
| | - Zhi Qian Hu
- Department of Surgery, Shanghai Chang Zheng Hospital, Second Military Medical University, Shanghai 200003, PR China
| | - Neill J Turner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Shi Feng Teng
- Department of Surgery, Shanghai Chang Zheng Hospital, Second Military Medical University, Shanghai 200003, PR China
| | - Wen Yue Cheng
- Department of Regenerative Medicine, Shanghai Zhabei District Central Hospital, Shanghai 200072, PR China
| | - Hai Yang Zhou
- Department of Surgery, Shanghai Chang Zheng Hospital, Second Military Medical University, Shanghai 200003, PR China
| | - Li Zhang
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Hong Wei Hu
- Department of General Surgery, Shanghai Zhabei District Central Hospital, Shanghai 200072, PR China
| | - Qiang Wang
- Department of Surgery, Shanghai Chang Zheng Hospital, Second Military Medical University, Shanghai 200003, PR China; Department of Regenerative Medicine, Shanghai Zhabei District Central Hospital, Shanghai 200072, PR China; Department of General Surgery, Shanghai Zhabei District Central Hospital, Shanghai 200072, PR China.
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| |
Collapse
|
109
|
Park KM, Hussein KH, Hong SH, Ahn C, Yang SR, Park SM, Kweon OK, Kim BM, Woo HM. Decellularized Liver Extracellular Matrix as Promising Tools for Transplantable Bioengineered Liver Promotes Hepatic Lineage Commitments of Induced Pluripotent Stem Cells. Tissue Eng Part A 2016; 22:449-60. [PMID: 26801816 DOI: 10.1089/ten.tea.2015.0313] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Liver transplantation is the last resort for liver failure patients. However, due to the shortage of donor organs, bioengineered liver generated from decellularized whole liver scaffolds and induced pluripotent stem cell (iPSC)-derived hepatocytes (iPSC-Heps) is being studied as an alternative approach to treat liver disease. Nevertheless, there has been no report on both the interaction of iPSC-Heps with a liver extracellular matrix (ECM) and the analysis of recellularized iPSC-Heps into the whole liver scaffolds. In this study, we produced porcine iPSC-Heps, which strongly expressed the hepatic markers α-fetoprotein and albumin and exhibited hepatic functionalities, including glycogen storage, lipid accumulation, low-density lipoprotein uptake, and indocyanine green metabolism. Supplementation of ECM from porcine decellularized liver containing liver-derived growth factors stimulated the albumin expression of porcine iPSC-Heps during differentiation procedures. The iPSC-Heps were reseeded into decellularized liver scaffolds, and the recellularized liver was cultured using a continuous perfusion system. The recellularized liver scaffolds were transplanted into rats for a short term, and the grafts expressed hepatocyte markers and did not rupture. These results provide a foundation for development of bioengineered liver using stem cell and decellularized scaffolds.
Collapse
Affiliation(s)
- Kyung-Mee Park
- 1 Stem Cell Institute-KNU, Kangwon National University , Chuncheon, Korea.,2 School of Veterinary Medicine, Kangwon National University , Chuncheon, Korea
| | - Kamal Hany Hussein
- 1 Stem Cell Institute-KNU, Kangwon National University , Chuncheon, Korea.,2 School of Veterinary Medicine, Kangwon National University , Chuncheon, Korea
| | - Seok-Ho Hong
- 1 Stem Cell Institute-KNU, Kangwon National University , Chuncheon, Korea.,3 School of Medicine, Kangwon National University , Chuncheon, Korea
| | - Cheol Ahn
- 1 Stem Cell Institute-KNU, Kangwon National University , Chuncheon, Korea.,4 School of Biomedical Technology, Kangwon National University , Chuncheon, Korea
| | - Se-Ran Yang
- 1 Stem Cell Institute-KNU, Kangwon National University , Chuncheon, Korea.,3 School of Medicine, Kangwon National University , Chuncheon, Korea
| | - Sung-Min Park
- 1 Stem Cell Institute-KNU, Kangwon National University , Chuncheon, Korea.,3 School of Medicine, Kangwon National University , Chuncheon, Korea
| | - Oh-Kyeong Kweon
- 5 School of Veterinary Medicine, Seoul National University , Seoul, Korea
| | - Byeong-Moo Kim
- 6 Departments of Medicine/GI Unit, Massachusetts General Hospital , Harvard Medical School, Boston, Massachusetts
| | - Heung-Myong Woo
- 1 Stem Cell Institute-KNU, Kangwon National University , Chuncheon, Korea.,2 School of Veterinary Medicine, Kangwon National University , Chuncheon, Korea
| |
Collapse
|
110
|
Poornejad N, Frost TS, Scott DR, Elton BB, Reynolds PR, Roeder BL, Cook AD. Freezing/Thawing without Cryoprotectant Damages Native but not Decellularized Porcine Renal Tissue. Organogenesis 2016; 11:30-45. [PMID: 25730294 DOI: 10.1080/15476278.2015.1022009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Whole organ decellularization of porcine renal tissue and recellularization with a patient's own cells would potentially overcome immunorejection, which is one of the most significant problems with allogeneic kidney transplantation. However, there are obstacles to achieving this goal, including preservation of the decellularized extracellular matrix (ECM), identifying the proper cell types, and repopulating the ECM before transplantation. Freezing biological tissue is the best option to avoid spoilage; however, it may damage the structure of the tissue or disrupt cellular membranes through ice crystal formation. Cryoprotectants have been used to repress ice formation during freezing, although cell toxicity can still occur. The effect of freezing/thawing on native (n = 10) and decellularized (n = 10) whole porcine kidneys was studied without using cryoprotectants. Results showed that the elastic modulus of native kidneys was reduced by a factor of 22 (P < 0.0001) by freezing/thawing or decellularization, while the elastic modulus for decellularized ECM was essentially unchanged by the freezing/thawing process (p = 0.0636). Arterial pressure, representative of structural integrity, was also reduced by a factor of 52 (P < 0.0001) after freezing/thawing for native kidneys, compared to a factor of 43 (P < 0.0001) for decellularization and a factor of 4 (P < 0.0001) for freezing/thawing decellularized structures. Both freezing/thawing and decellularization reduced stiffness, but the reductions were not additive. Investigation of the microstructure of frozen/thawed native and decellularized renal tissues showed increased porosity due to cell removal and ice crystal formation. Orcein and Sirius staining showed partial damage to elastic and collagen fibers after freezing/thawing. It was concluded that cellular damage and removal was more responsible for reducing stiffness than fibril destruction. Cell viability and growth were demonstrated on decellularized frozen/thawed and non-frozen samples using human renal cortical tubular epithelial (RCTE) cells over 12 d. No adverse effect on the ability to recellularize after freezing/thawing was observed. It is recommended that porcine kidneys be frozen prior to decellularization to prevent contamination, and after decellularization to prevent protein denaturation. Cryoprotectants may still be necessary, however, during storage and transportation after recellularization.
Collapse
Affiliation(s)
- Nafiseh Poornejad
- a Department of Chemical Engineering; Brigham Young University ; Provo , UT , USA
| | | | | | | | | | | | | |
Collapse
|
111
|
Lin YQ, Wang LR, Pan LL, Wang H, Zhu GQ, Liu WY, Wang JT, Braddock M, Zheng MH. Kidney bioengineering in regenerative medicine: An emerging therapy for kidney disease. Cytotherapy 2016; 18:186-197. [PMID: 26596504 DOI: 10.1016/j.jcyt.2015.10.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 09/21/2015] [Accepted: 10/05/2015] [Indexed: 12/21/2022]
Abstract
The prevalence of end-stage renal disease is emerging as a serious worldwide public health problem because of the shortage of donor organs and the need to take lifelong immunosuppressive medication in patients who receive a transplanted kidney. Recently, tissue bioengineering of decellularization and recellularization scaffolds has emerged as a novel strategy for organ regeneration, and we review the critical technologies supporting these methods. We present a summary of factors associated with experimental protocols that may shed light on the future development of kidney bioengineering and we discuss the cell sources and bioreactor techniques applied to the recellularization process. Finally, we review some artificial renal engineering technologies and their future prospects, such as kidney on a chip and the application of three-dimensional and four-dimensional printing in kidney tissue engineering.
Collapse
Affiliation(s)
- Yi-Qian Lin
- Department of Infection and Liver Diseases, Liver Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Renji School of Wenzhou Medical University, Wenzhou, China
| | - Li-Ren Wang
- Department of Infection and Liver Diseases, Liver Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Liang-Liang Pan
- School of Laboratory and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Hui Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gui-Qi Zhu
- Department of Infection and Liver Diseases, Liver Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wen-Yue Liu
- Department of Endocrinology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiang-Tao Wang
- Department of Infection and Liver Diseases, Liver Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Martin Braddock
- Global Medicines Development, AstraZeneca R&D, Alderley Park, United Kingdom
| | - Ming-Hua Zheng
- Department of Infection and Liver Diseases, Liver Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Institute of Hepatology, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
112
|
Petrosyan A, Zanusso I, Lavarreda-Pearce M, Leslie S, Sedrakyan S, De Filippo RE, Orlando G, Da Sacco S, Perin L. Decellularized Renal Matrix and Regenerative Medicine of the Kidney: A Different Point of View. TISSUE ENGINEERING PART B-REVIEWS 2016; 22:183-92. [PMID: 26653996 DOI: 10.1089/ten.teb.2015.0368] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Over the past years, extracellular matrix (ECM) obtained from whole organ decellularization has been investigated as a platform for organ engineering. The ECM is composed of fibrous and nonfibrous molecules providing structural and biochemical support to the surrounding cells. Multiple decellularization techniques, including ours, have been optimized to maintain the composition, microstructure, and biomechanical properties of the native renal ECM that are difficult to obtain during the generation of synthetic substrates. There are evidences suggesting that in vivo implanted renal ECM has the capacity to induce formation of vasculature-like structures, but long-term in vivo transplantation and filtration activity by these tissue-engineered constructs have not been investigated or reported. Therefore, even if the process of renal decellularization is possible, the repopulation of the renal matrix with functional renal cell types is still very challenging. This review aims to summarize the current reports on kidney tissue engineering with the use of decellularized matrices and addresses the challenges in creating functional kidney units. Finally, this review discusses how future studies investigating cell-matrix interaction may aid the generation of a functional renal unit that would be transplantable into patients one day.
Collapse
Affiliation(s)
- Astgik Petrosyan
- 1 Department of Development, Stem Cells and Regenerative Medicine, University of Southern California , Los Angeles, California
| | - Ilenia Zanusso
- 2 Department of Urology, Children's Hospital Los Angeles , Los Angeles, California
| | | | - Scott Leslie
- 2 Department of Urology, Children's Hospital Los Angeles , Los Angeles, California
| | - Sargis Sedrakyan
- 2 Department of Urology, Children's Hospital Los Angeles , Los Angeles, California
| | - Roger E De Filippo
- 2 Department of Urology, Children's Hospital Los Angeles , Los Angeles, California
| | - Giuseppe Orlando
- 3 Department of General Surgery, Wake Forest School of Medicine , Winston Salem, North Carolina
| | - Stefano Da Sacco
- 2 Department of Urology, Children's Hospital Los Angeles , Los Angeles, California
| | - Laura Perin
- 2 Department of Urology, Children's Hospital Los Angeles , Los Angeles, California
| |
Collapse
|
113
|
Jin M, Yaling Y, Zhibin W, Jianse Z. Decellularization of Rat Kidneys to Produce Extracellular Matrix Scaffolds. Methods Mol Biol 2016; 1397:53-63. [PMID: 26676127 DOI: 10.1007/978-1-4939-3353-2_6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The extracellular matrix (ECM) retains three-dimensional structures for the stimulation of cell growth, with components of the ECM relatively conserved between species. Interest in the use of decellularized scaffold-based strategies for organ regeneration is increasing rapidly. Decellularized scaffolds derived from animal organs are a promising material for organ engineering, with a number of prominent advances having been reported in the past few years.In this article we describe a simple and robust methodology for generating decellularized rat kidneys. To obtain these scaffolds, we perfuse rat kidneys with detergents through the abdominal aorta. After decellularization, kidney scaffolds are harvested for evaluation of vascular structure and histology. Qualitative evaluation involves vascular corrosion casting, transmission electron microscopy, and several different histological and immunofluorescent methods. SDS residue levels are assessed by ultraviolet-visible spectrophotometer (UV-VIS).
Collapse
Affiliation(s)
- Mei Jin
- Anatomy Department & Institute of Bioscaffold Transplantation and Immunology, Wenzhou Medical University, 1210 Wenzhou University Town, Wenzhou, Zhejiang, 325035, China.
| | - Yu Yaling
- Anatomy Department & Institute of Bioscaffold Transplantation and Immunology, Wenzhou Medical University, 1210 Wenzhou University Town, Wenzhou, Zhejiang, 325035, China
| | - Wang Zhibin
- Anatomy Department & Institute of Bioscaffold Transplantation and Immunology, Wenzhou Medical University, 1210 Wenzhou University Town, Wenzhou, Zhejiang, 325035, China
| | - Zhang Jianse
- Anatomy Department & Institute of Bioscaffold Transplantation and Immunology, Wenzhou Medical University, 1210 Wenzhou University Town, Wenzhou, Zhejiang, 325035, China
| |
Collapse
|
114
|
Batchelder CA, Martinez ML, Tarantal AF. Natural Scaffolds for Renal Differentiation of Human Embryonic Stem Cells for Kidney Tissue Engineering. PLoS One 2015; 10:e0143849. [PMID: 26645109 PMCID: PMC4672934 DOI: 10.1371/journal.pone.0143849] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 11/09/2015] [Indexed: 01/01/2023] Open
Abstract
Despite the enthusiasm for bioengineering of functional renal tissues for transplantation, many obstacles remain before the potential of this technology can be realized in a clinical setting. Viable tissue engineering strategies for the kidney require identification of the necessary cell populations, efficient scaffolds, and the 3D culture conditions to develop and support the unique architecture and physiological function of this vital organ. Our studies have previously demonstrated that decellularized sections of rhesus monkey kidneys of all age groups provide a natural extracellular matrix (ECM) with sufficient structural properties with spatial and organizational influences on human embryonic stem cell (hESC) migration and differentiation. To further explore the use of decellularized natural kidney scaffolds for renal tissue engineering, pluripotent hESC were seeded in whole- or on sections of kidney ECM and cell migration and phenotype compared with the established differentiation assays for hESC. Results of qPCR and immunohistochemical analyses demonstrated upregulation of renal lineage markers when hESC were cultured in decellularized scaffolds without cytokine or growth factor stimulation, suggesting a role for the ECM in directing renal lineage differentiation. hESC were also differentiated with growth factors and compared when seeded on renal ECM or a new biologically inert polysaccharide scaffold for further maturation. Renal lineage markers were progressively upregulated over time on both scaffolds and hESC were shown to express signature genes of renal progenitor, proximal tubule, endothelial, and collecting duct populations. These findings suggest that natural scaffolds enhance expression of renal lineage markers particularly when compared to embryoid body culture. The results of these studies show the capabilities of a novel polysaccharide scaffold to aid in defining a protocol for renal progenitor differentiation from hESC, and advance the promise of tissue engineering as a source of functional kidney tissue.
Collapse
Affiliation(s)
- Cynthia A. Batchelder
- California National Primate Research Center, University of California, Davis, California, United States of America
| | - Michele L. Martinez
- California National Primate Research Center, University of California, Davis, California, United States of America
| | - Alice F. Tarantal
- California National Primate Research Center, University of California, Davis, California, United States of America
- Department of Pediatrics, School of Medicine, University of California, Davis, California, United States of America
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
115
|
Rogers J, Katari R, Gifford S, Tamburrini R, Edgar L, Voigt MR, Murphy SV, Igel D, Mancone S, Callese T, Colucci N, Mirzazadeh M, Peloso A, Zambon JP, Farney AC, Stratta RJ, Orlando G. Kidney transplantation, bioengineering and regeneration: an originally immunology-based discipline destined to transition towards ad hoc organ manufacturing and repair. Expert Rev Clin Immunol 2015; 12:169-82. [PMID: 26634874 DOI: 10.1586/1744666x.2016.1112268] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Kidney transplantation (KT), as a modality of renal replacement therapy (RRT), has been shown to be both economically and functionally superior to dialysis for the treatment of end-stage renal disease (ESRD). Progress in KT is limited by two major barriers: a) a chronic and burgeoning shortage of transplantable organs and b) the need for chronic immunosuppression following transplantation. Although ground-breaking advances in transplant immunology have improved patient survival and graft durability, a new pathway of innovation is needed in order to overcome current obstacles. Regenerative medicine (RM) holds the potential to shift the paradigm in RRT, through organ bioengineering. Manufactured organs represent a potentially inexhaustible source of transplantable grafts that would bypass the need for immunosuppressive drugs by using autologous cells to repopulate extracellular matrix (ECM) scaffolds. This overview discusses the current status of renal transplantation while reviewing the most promising innovations in RM therapy as applied to RRT.
Collapse
Affiliation(s)
- Jeffrey Rogers
- a Department of Surgery , Wake Forest University , Winston Salem , NC , USA
| | - Ravi Katari
- b Wake Forest University School of Medicine , Winston Salem , NC , USA
| | - Sheyna Gifford
- c Annenberg School for Communication & Journalism , University of Southern California , Los Angeles , CA , USA
| | | | - Lauren Edgar
- b Wake Forest University School of Medicine , Winston Salem , NC , USA
| | - Marcia R Voigt
- b Wake Forest University School of Medicine , Winston Salem , NC , USA
| | - Sean V Murphy
- d Wake Forest Institute for Regenerative Medicine , Winston Salem , NC , USA
| | - Daniel Igel
- b Wake Forest University School of Medicine , Winston Salem , NC , USA
| | - Sara Mancone
- b Wake Forest University School of Medicine , Winston Salem , NC , USA
| | - Tyler Callese
- b Wake Forest University School of Medicine , Winston Salem , NC , USA
| | - Nicola Colucci
- a Department of Surgery , Wake Forest University , Winston Salem , NC , USA
| | - Majid Mirzazadeh
- e Department of Urology , Wake Forest University , Winston Salem , NC , USA
| | - Andrea Peloso
- f Department of General Surgery , University of Pavia , Pavia , Italy
| | - Joao Paulo Zambon
- d Wake Forest Institute for Regenerative Medicine , Winston Salem , NC , USA
| | - Alan C Farney
- a Department of Surgery , Wake Forest University , Winston Salem , NC , USA
| | - Robert J Stratta
- a Department of Surgery , Wake Forest University , Winston Salem , NC , USA
| | - Giuseppe Orlando
- a Department of Surgery , Wake Forest University , Winston Salem , NC , USA
| |
Collapse
|
116
|
Zhou P, Huang Y, Guo Y, Wang L, Ling C, Guo Q, Wang Y, Zhu S, Fan X, Zhu M, Huang H, Lu Y, Wang Z. Decellularization and Recellularization of Rat Livers With Hepatocytes and Endothelial Progenitor Cells. Artif Organs 2015; 40:E25-38. [PMID: 26637111 DOI: 10.1111/aor.12645] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Pengcheng Zhou
- Department of General Surgery; Affiliated Hospital of Nantong University; Nantong China
- Department of Emergency Surgery; Affiliated Hospital of Nantong University; Nantong China
| | - Yan Huang
- Department of General Surgery; Affiliated Hospital of Nantong University; Nantong China
| | - Yibing Guo
- Surgical Comprehensive Laboratory; Affiliated Hospital of Nantong University; Nantong China
| | - Lei Wang
- Department of General Surgery; Affiliated Hospital of Nantong University; Nantong China
| | - Changchun Ling
- Department of General Surgery; Affiliated Hospital of Nantong University; Nantong China
| | - Qingsong Guo
- Department of General Surgery; Affiliated Hospital of Nantong University; Nantong China
| | - Yao Wang
- Department of General Surgery; Affiliated Hospital of Nantong University; Nantong China
| | - Shajun Zhu
- Department of General Surgery; Affiliated Hospital of Nantong University; Nantong China
| | - Xiangjun Fan
- Department of General Surgery; Affiliated Hospital of Nantong University; Nantong China
| | - Mingyan Zhu
- Department of General Surgery; Affiliated Hospital of Nantong University; Nantong China
| | - Hua Huang
- Department of Pathology; Affiliated Hospital of Nantong University; Nantong China
| | - Yuhua Lu
- Department of General Surgery; Affiliated Hospital of Nantong University; Nantong China
- Surgical Comprehensive Laboratory; Affiliated Hospital of Nantong University; Nantong China
| | - Zhiwei Wang
- Department of General Surgery; Affiliated Hospital of Nantong University; Nantong China
| |
Collapse
|
117
|
Poornejad N, Nielsen JJ, Morris RJ, Gassman JR, Reynolds PR, Roeder BL, Cook AD. Comparison of four decontamination treatments on porcine renal decellularized extracellular matrix structure, composition, and support of human renal cortical tubular epithelium cells. J Biomater Appl 2015; 30:1154-67. [PMID: 26589294 DOI: 10.1177/0885328215615760] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Engineering whole organs from porcine decellularized extracellular matrix and human cells may lead to a plentiful source of implantable organs. Decontaminating the porcine decellularized extracellular matrix scaffolds is an essential step prior to introducing human cells. However, decontamination of whole porcine kidneys is a major challenge because the decontamination agent or irradiation needs to diffuse deep into the structure to eliminate all microbial contamination while minimizing damage to the structure and composition of the decellularized extracellular matrix. In this study, we compared four decontamination treatments that could be applicable to whole porcine kidneys: 70% ethanol, 0.2% peracetic acid in 1 M NaCl, 0.2% peracetic acid in 4% ethanol, and gamma (γ)-irradiation. Porcine kidneys were decellularized by perfusion of 0.5% (w/v) aqueous solution of sodium dodecyl sulfate and the four decontamination treatments were optimized using segments (n = 60) of renal tissue to ensure a consistent comparison. Although all four methods were successful in decontamination, γ-irradiation was very damaging to collagen fibers and glycosaminoglycans, leading to less proliferation of human renal cortical tubular epithelium cells within the porcine decellularized extracellular matrix. The effectiveness of the other three optimized solution treatments were then all confirmed using whole decellularized porcine kidneys (n = 3). An aqueous solution of 0.2% peracetic acid in 1 M NaCl was determined to be the best method for decontamination of porcine decellularized extracellular matrix.
Collapse
Affiliation(s)
- Nafiseh Poornejad
- Department of Chemical Engineering, Brigham Young University, Provo, UT, USA
| | - Jeffery J Nielsen
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Ryan J Morris
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT, USA
| | - Jason R Gassman
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT, USA
| | - Paul R Reynolds
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT, USA
| | | | - Alonzo D Cook
- Department of Chemical Engineering, Brigham Young University, Provo, UT, USA
| |
Collapse
|
118
|
Peloso A, Petrosyan A, Da Sacco S, Booth C, Zambon JP, OʼBrien T, Aardema C, Robertson J, De Filippo RE, Soker S, Stratta RJ, Perin L, Orlando G. Renal Extracellular Matrix Scaffolds From Discarded Kidneys Maintain Glomerular Morphometry and Vascular Resilience and Retains Critical Growth Factors. Transplantation 2015; 99:1807-16. [PMID: 26018349 DOI: 10.1097/tp.0000000000000811] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Extracellular matrix (ECM) scaffolds, obtained through detergent-based decellularization of native kidneys, represent the most promising platform for investigations aiming at manufacturing kidneys for transplant purposes. We previously showed that decellularization of the human kidney yields renal ECM scaffolds (hrECMs) that maintain their basic molecular components, are cytocompatible, stimulate angiogenesis, and show an intact innate vasculature. However, evidence that the decellularization preserves glomerular morphometric characteristics, physiological parameters (pressures and resistances of the vasculature bed), and biological properties of the renal ECM, including retention of important growth factors (GFs), is still missing. METHODS To address these issues, we studied the morphometry and resilience of hrECMs' native vasculature with resin casting at electronic microscopy and pulse-wave measurements, respectively. Moreover, we determined the fate of 40 critical GFs post decellularization with a glass chip-based multiplex enzyme-linked immunosorbent assay array and in vitro immunofluorescence. RESULTS Our method preserves the 3-dimensional conformation of the native glomerulus. Resin casting and pulse-wave measurements, showed that hrECMs preserves the microvascular morphology and morphometry, and physiological function. Moreover, GFs including vascular endothelial growth factor and its receptors are retained within the matrices. CONCLUSIONS Our results indicate that discarded human kidneys are a suitable source of renal scaffolds because they maintain a well-preserved structure and function of the vasculature, as well as GFs that are fundamental to achieve a satisfying recellularization of the scaffold in vivo due to their angiogenic properties.
Collapse
Affiliation(s)
- Andrea Peloso
- 1 Wake Forest School of Medicine, Winston Salem, NC. 2 General Surgery, Fondazione IRCCS Policlinico San Matteo Pavia and University of Pavia, Pavia, Italy. 3 GOFARR Laboratory, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA. 4 Department of Urology, University of Southern California, Los Angeles, CA. 5 Departments of Biomedical and Mechanical Engineering, Virginia Tech, Blacksburg, VA. 6 Smart Perfusion LLC, Denver, NC
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
119
|
Perfusion Decellularization of Discarded Human Kidneys: A Valuable Platform for Organ Regeneration. Transplantation 2015. [PMID: 26203856 DOI: 10.1097/tp.0000000000000810] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
120
|
Katari R, Edgar L, Wong T, Boey A, Mancone S, Igel D, Callese T, Voigt M, Tamburrini R, Zambon JP, Perin L, Orlando G. Tissue-Engineering Approaches to Restore Kidney Function. Curr Diab Rep 2015; 15:69. [PMID: 26275443 DOI: 10.1007/s11892-015-0643-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Kidney transplantation for the treatment of chronic kidney disease has established outcome and quality of life. However, its implementation is severely limited by a chronic shortage of donor organs; consequently, most candidates remain on dialysis and on the waiting list while accruing further morbidity and mortality. Furthermore, those patients that do receive kidney transplants are committed to a life-long regimen of immunosuppressive drugs that also carry significant adverse risk profiles. The disciplines of tissue engineering and regenerative medicine have the potential to produce alternative therapies which circumvent the obstacles posed by organ shortage and immunorejection. This review paper describes some of the most promising tissue-engineering solutions currently under investigation for the treatment of acute and chronic kidney diseases. The various stem cell therapies, whole embryo transplantation, and bioengineering with ECM scaffolds are outlined and summarized.
Collapse
Affiliation(s)
- Ravi Katari
- Section of Transplantation, Department of Surgery, Wake Forest School of Medicine, Winston Salem, NC, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
121
|
Abstract
PURPOSE OF REVIEW Renal transplantation is currently the only definitive treatment for end-stage renal disease; however, this treatment is severely limited by the shortage of implantable kidneys. To address this shortcoming, development of an engineered, transplantable kidney has been proposed. Although current advances in engineering kidneys based on decellularization and recellularization techniques have offered great promises for the generation of functional kidney constructs, most studies have been conducted using rodent kidney constructs and short-term in-vivo evaluation. Toward clinical translations of this technique, several limitations need to be addressed. RECENT FINDINGS Human-sized renal scaffolds are desirable for clinical application, and the fabrication is currently feasible using native porcine and discarded human kidneys. Current progress in stem cell biology and cell culture methods have demonstrated feasibility of the use of embryonic stem cells, induced pluripotent stem cells, and primary renal cells as clinically relevant cell sources for the recellularization of renal scaffolds. Finally, approaches to long-term implantation of engineered kidneys are under investigation using antithrombogenic strategies such as functional reendothelialization of acellular kidney matrices. SUMMARY In the field of bioengineering, whole kidneys have taken a number of important initial steps toward clinical translations, but many challenges must be addressed to achieve a successful treatment for the patient with end-stage renal disease.
Collapse
|
122
|
Prakash YS, Tschumperlin DJ, Stenmark KR. Coming to terms with tissue engineering and regenerative medicine in the lung. Am J Physiol Lung Cell Mol Physiol 2015; 309:L625-38. [PMID: 26254424 DOI: 10.1152/ajplung.00204.2015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/04/2015] [Indexed: 01/10/2023] Open
Abstract
Lung diseases such as emphysema, interstitial fibrosis, and pulmonary vascular diseases cause significant morbidity and mortality, but despite substantial mechanistic understanding, clinical management options for them are limited, with lung transplantation being implemented at end stages. However, limited donor lung availability, graft rejection, and long-term problems after transplantation are major hurdles to lung transplantation being a panacea. Bioengineering the lung is an exciting and emerging solution that has the ultimate aim of generating lung tissues and organs for transplantation. In this article we capture and review the current state of the art in lung bioengineering, from the multimodal approaches, to creating anatomically appropriate lung scaffolds that can be recellularized to eventually yield functioning, transplant-ready lungs. Strategies for decellularizing mammalian lungs to create scaffolds with native extracellular matrix components vs. de novo generation of scaffolds using biocompatible materials are discussed. Strengths vs. limitations of recellularization using different cell types of various pluripotency such as embryonic, mesenchymal, and induced pluripotent stem cells are highlighted. Current hurdles to guide future research toward achieving the clinical goal of transplantation of a bioengineered lung are discussed.
Collapse
Affiliation(s)
- Y S Prakash
- Department of Anesthesiology, Mayo Clinic, Rochester, Minnesota; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota;
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota; Division of Pulmonary Medicine, Mayo Clinic, Rochester, Minnesota; and
| | - Kurt R Stenmark
- Department of Pediatrics, University of Colorado, Aurora, Colorado
| |
Collapse
|
123
|
Kawasaki T, Kirita Y, Kami D, Kitani T, Ozaki C, Itakura Y, Toyoda M, Gojo S. Novel detergent for whole organ tissue engineering. J Biomed Mater Res A 2015; 103:3364-73. [PMID: 25850947 DOI: 10.1002/jbm.a.35474] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/17/2015] [Accepted: 04/02/2015] [Indexed: 02/03/2023]
Abstract
Whole organ tissue engineering for various organs, including the heart, lung, liver, and kidney, has demonstrated promising results for end-stage organ failure. However, the sodium dodecyl sulfate (SDS)-based protocol for standard decellularization has drawbacks such as clot formation in vascularized transplantation and poor cell engraftment in recellularization procedures. Preservation of the surface milieu of extracellular matrices (ECMs) might be crucial for organ generation based on decellularization/recellularization engineering. We examined a novel detergent, sodium lauryl ether sulfate (SLES), to determine whether it could overcome the drawbacks associated with SDS using rat heart and kidney. Both organs were perfused in an antegrade fashion with either SLES or SDS. Although immunohistochemistry for collagen I, IV, laminin, and fibronectin showed similar preservation in both detergents, morphological analysis using scanning electron microscopy and an assay of glycosaminoglycan content on ECMs showed that SLES-treated tissues had better-preserved ECMs than SDS-treated tissues. Mesenteric transplantation revealed SLES did not induce significant inflammation, as opposed to SDS. Platelet adhesion to decellularized tissues was significantly reduced with SLES. Overall, SLES could replace older detergents such as SDS in the decellularization process for generation of transplantable recellularized organs.
Collapse
Affiliation(s)
- Takanori Kawasaki
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kamigyo ku, Kyoto, 602-8566, Japan
| | - Yuhei Kirita
- Department of Nephrology, Kyoto Prefectural University of Medicine, Kamigyo ku, Kyoto, 602-8566, Japan
| | - Daisuke Kami
- Department of Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo ku, Kyoto, 602-8566, Japan
| | - Tomoya Kitani
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kamigyo ku, Kyoto, 602-8566, Japan
| | - Chisa Ozaki
- Sanyo Chemical Industries, Ltd, Biomedical Product Itakura, Yako
| | - Yoko Itakura
- Department of Vascular Medicine, Tokyo Metropolitan Institute of Gerontology, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Masashi Toyoda
- Department of Vascular Medicine, Tokyo Metropolitan Institute of Gerontology, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Satoshi Gojo
- Department of Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo ku, Kyoto, 602-8566, Japan
| |
Collapse
|
124
|
Bao J, Wu Q, Sun J, Zhou Y, Wang Y, Jiang X, Li L, Shi Y, Bu H. Hemocompatibility improvement of perfusion-decellularized clinical-scale liver scaffold through heparin immobilization. Sci Rep 2015; 5:10756. [PMID: 26030843 PMCID: PMC5377232 DOI: 10.1038/srep10756] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 04/27/2015] [Indexed: 02/05/2023] Open
Abstract
Whole-liver perfusion-decellularization is an attractive scaffold-preparation technique for producing clinical transplantable liver tissue. However, the scaffold's poor hemocompatibility poses a major obstacle. This study was intended to improve the hemocompatibility of perfusion-decellularized porcine liver scaffold via immobilization of heparin. Heparin was immobilized on decellularized liver scaffolds (DLSs) by electrostatic binding using a layer-by-layer self-assembly technique (/h-LBL scaffold), covalent binding via multi-point attachment (/h-MPA scaffold), or end-point attachment (/h-EPA scaffold). The effect of heparinization on anticoagulant ability and cytocompatibility were investigated. The result of heparin content and release tests revealed EPA technique performed higher efficiency of heparin immobilization than other two methods. Then, systematic in vitro investigation of prothrombin time (PT), thrombin time (TT), activated partial thromboplastin time (APTT), platelet adhesion and human platelet factor 4 (PF4, indicates platelet activation) confirmed the heparinized scaffolds, especially the /h-EPA counterparts, exhibited ultralow blood component activations and excellent hemocompatibility. Furthermore, heparin treatments prevented thrombosis successfully in DLSs with blood perfusion after implanted in vivo. Meanwhile, after heparin processes, both primary hepatocyte and endothelial cell viability were also well-maintained, which indicated that heparin treatments with improved biocompatibility might extend to various hemoperfusable whole-organ scaffolds' preparation.
Collapse
Affiliation(s)
- Ji Bao
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiong Wu
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiu Sun
- Department of General Surgery, The first people’s hospital of Yibin, Yibin, 644000, China
| | - Yongjie Zhou
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yujia Wang
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xin Jiang
- College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, China
| | - Li Li
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yujun Shi
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hong Bu
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
125
|
da Palma RK, Campillo N, Uriarte JJ, Oliveira LVF, Navajas D, Farré R. Pressure- and flow-controlled media perfusion differently modify vascular mechanics in lung decellularization. J Mech Behav Biomed Mater 2015; 49:69-79. [PMID: 26002417 DOI: 10.1016/j.jmbbm.2015.04.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 04/18/2015] [Accepted: 04/23/2015] [Indexed: 01/07/2023]
Abstract
Organ biofabrication is a potential future alternative for obtaining viable organs for transplantation. Achieving intact scaffolds to be recellularized is a key step in lung bioengineering. Perfusion of decellularizing media through the pulmonary artery has shown to be effective. How vascular perfusion pressure and flow vary throughout lung decellularization, which is not well known, is important for optimizing the process (minimizing time) while ensuring scaffold integrity (no barotrauma). This work was aimed at characterizing the pressure/flow relationship at the pulmonary vasculature and at how effective vascular resistance depends on pressure- and flow-controlled variables when applying different methods of media perfusion for lung decellularization. Lungs from 43 healthy mice (C57BL/6; 7-8 weeks old) were investigated. After excision and tracheal cannulation, lungs were inflated at 10 cmH2O airway pressure and subjected to conventional decellularization with a solution of 1% sodium dodecyl sulfate (SDS). Pressure (PPA) and flow (V'PA) at the pulmonary artery were continuously measured. Decellularization media was perfused through the pulmonary artery: (a) at constant PPA=20 cmH2O or (b) at constant V'PA=0.5 and 0.2 ml/min. Effective vascular resistance was computed as Rv=PPA/V'PA. Rv (in cmH2O/(ml/min)); mean±SE) considerably varied throughout lung decellularization, particularly for pressure-controlled perfusion (from 29.1±3.0 in baseline to a maximum of 664.1±164.3 (p<0.05), as compared with flow-controlled perfusion (from 49.9±3.3 and 79.5±5.1 in baseline to a maximum of 114.4±13.9 and 211.7±70.5 (p<0.05, both), for V'PA of 0.5 and 0.2 ml/min respectively. Most of the media infused to the pulmonary artery throughout decellularization circulated to the airways compartment across the alveolar-capillary membrane. This study shows that monitoring perfusion mechanics throughout decellularization provides information relevant for optimizing the process time while ensuring that vascular pressure is kept within a safety range to preserve the organ scaffold integrity.
Collapse
Affiliation(s)
- Renata K da Palma
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Master's and Doctoral Degree Programs in Rehabilitation Sciences, Nove de Julho University, Sao Paulo, Brazil
| | - Noelia Campillo
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Institut de Bioenginyeria de Catalunya, Barcelona, Spain
| | - Juan J Uriarte
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; CIBER Enfermedades Respiratorias, Madrid, Spain
| | - Luis V F Oliveira
- Master's and Doctoral Degree Programs in Rehabilitation Sciences, Nove de Julho University, Sao Paulo, Brazil
| | - Daniel Navajas
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Institut de Bioenginyeria de Catalunya, Barcelona, Spain; CIBER Enfermedades Respiratorias, Madrid, Spain
| | - Ramon Farré
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; CIBER Enfermedades Respiratorias, Madrid, Spain; Institut Investigacions Biomediques August Pi Sunyer, Barcelona, Spain.
| |
Collapse
|
126
|
Paulsen SJ, Miller JS. Tissue vascularization through 3D printing: Will technology bring us flow? Dev Dyn 2015; 244:629-40. [PMID: 25613150 DOI: 10.1002/dvdy.24254] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 01/08/2015] [Accepted: 01/13/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Though in vivo models provide the most physiologically relevant environment for studying tissue function, in vitro studies provide researchers with explicit control over experimental conditions and the potential to develop high throughput testing methods. In recent years, advancements in developmental biology research and imaging techniques have significantly improved our understanding of the processes involved in vascular development. However, the task of recreating the complex, multi-scale vasculature seen in in vivo systems remains elusive. RESULTS 3D bioprinting offers a potential method to generate controlled vascular networks with hierarchical structure approaching that of in vivo networks. Bioprinting is an interdisciplinary field that relies on advances in 3D printing technology along with advances in imaging and computational modeling, which allow researchers to monitor cellular function and to better understand cellular environment within the printed tissue. CONCLUSIONS As bioprinting technologies improve with regards to resolution, printing speed, available materials, and automation, 3D printing could be used to generate highly controlled vascularized tissues in a high throughput manner for use in regenerative medicine and the development of in vitro tissue models for research in developmental biology and vascular diseases.
Collapse
Affiliation(s)
- S J Paulsen
- Department of Bioengineering, Rice University, Houston, Texas
| | | |
Collapse
|
127
|
Scarritt ME, Pashos NC, Bunnell BA. A review of cellularization strategies for tissue engineering of whole organs. Front Bioeng Biotechnol 2015; 3:43. [PMID: 25870857 PMCID: PMC4378188 DOI: 10.3389/fbioe.2015.00043] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/16/2015] [Indexed: 12/22/2022] Open
Abstract
With the advent of whole organ decellularization, extracellular matrix scaffolds suitable for organ engineering were generated from numerous tissues, including the heart, lung, liver, kidney, and pancreas, for use as alternatives to traditional organ transplantation. Biomedical researchers now face the challenge of adequately and efficiently recellularizing these organ scaffolds. Herein, an overview of whole organ decellularization and a thorough review of the current literature for whole organ recellularization are presented. The cell types, delivery methods, and bioreactors employed for recellularization are discussed along with commercial and clinical considerations, such as immunogenicity, biocompatibility, and Food and Drug Administartion regulation.
Collapse
Affiliation(s)
- Michelle E Scarritt
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine , New Orleans, LA , USA
| | - Nicholas C Pashos
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine , New Orleans, LA , USA ; Bioinnovation PhD Program, Tulane University , New Orleans, LA , USA
| | - Bruce A Bunnell
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine , New Orleans, LA , USA ; Department of Pharmacology, Tulane University School of Medicine , New Orleans, LA , USA
| |
Collapse
|
128
|
Abstract
PURPOSE OF REVIEW The severe shortage of suitable donor kidneys limits organ transplantation to a small fraction of patients suffering from end-stage renal failure. Engineering autologous kidney grafts on-demand would potentially alleviate this shortage, thereby reducing healthcare costs, improving quality of life, and increasing longevity for patients suffering from renal failure. RECENT FINDINGS Over the past 2 years, several studies have demonstrated that structurally intact extracellular matrix (ECM) scaffolds can be derived from human or animal kidneys through decellularization, a process in which detergent or enzyme solutions are perfused through the renal vasculature to remove the native cells. The future clinical paradigm would be to repopulate these decellularized kidney matrices with patient-derived renal stem cells to regenerate a functional kidney graft. Recent research aiming toward this goal has focused on the optimization of decellularization protocols, design of bioreactor systems to seed cells into appropriate compartments of the renal ECM to nurture their growth to restore kidney function, and differentiation of pluripotent stem cells (PSCs) into renal progenitor lineages. SUMMARY New research efforts utilizing bio-mimetic perfusion bioreactor systems to repopulate decellularized kidney scaffolds, coupled with the differentiation of PSCs into renal progenitor cell populations, indicate substantial progress toward the ultimate goal of building a functional kidney graft on-demand.
Collapse
|
129
|
Scaffolds from surgically removed kidneys as a potential source of organ transplantation. BIOMED RESEARCH INTERNATIONAL 2015; 2015:325029. [PMID: 25756044 PMCID: PMC4338377 DOI: 10.1155/2015/325029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 01/18/2015] [Accepted: 01/18/2015] [Indexed: 01/07/2023]
Abstract
End stage renal disease (ESRD) is a common disease, which relates to nearly 600 million people in the total population. What is more, it seems to be a crucial problem from the epidemiological point of view. These facts lead to a further necessity of renal replacement therapy development connected with rising expenditures for the health care system. The aim of kidney tissue engineering is to develop and innovate methods of obtaining renal extracellular matrix (ECM) scaffolds derived from kidney decellularization. Recently, progress has been made towards developing a functional kidney graft in vitro on demand. In fact, decellularized tissues constitute ideal natural scaffolds, due to the preservation of native ECM architecture, as well as of cell-ECM binding domains critical in promoting cell attachment, migration, and proliferation. One of the potential sources of the natural scaffolds is the kidney, which cannot be transplanted immediately after excision.
Collapse
|
130
|
Peloso A, Katari R, Murphy SV, Zambon JP, DeFrancesco A, Farney AC, Rogers J, Stratta RJ, Manzia TM, Orlando G. Prospect for kidney bioengineering: shortcomings of the status quo. Expert Opin Biol Ther 2015; 15:547-58. [PMID: 25640286 DOI: 10.1517/14712598.2015.993376] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Dialysis and renal transplantation are the only two therapeutic options offered to patients affected by end-stage kidney disease; however, neither treatment can be considered definitive. In fact, dialysis is able to replace only the filtration function of the kidney without substituting its endocrine and metabolic roles, and dramatically impacts on patient's quality of life. On the other hand, kidney transplantation is severely limited by the shortage of transplantable organs, the need for immunosuppressive therapies and a narrow half-life. Regenerative medicine approaches are promising tools aiming to improve this condition. AREAS COVERED Cell therapies, bioartificial kidney, organ bioengineering, 3D printer and kidney-on-chip represent the most appealing areas of research for the treatment of end-stage kidney failure. The scope of this review is to summarize the state of the art, limits and directions of each branch. EXPERT OPINION In the future, these emerging technologies could provide definitive, curative and theoretically infinite options for the treatment of end-stage kidney disease. Progress in stem cells-based therapies, decellularization techniques and the more recent scientific know-how for the use of the 3D printer and kidney-on-chip could lead to a perfect cellular-based therapy, the futuristic creation of a bioengineered kidney in the lab or to a valid bioartificial alternative.
Collapse
Affiliation(s)
- Andrea Peloso
- Wake Forest School of Medicine , Winston-Salem, NC , USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Ko IK, Peng L, Peloso A, Smith CJ, Dhal A, Deegan DB, Zimmerman C, Clouse C, Zhao W, Shupe TD, Soker S, Yoo JJ, Atala A. Bioengineered transplantable porcine livers with re-endothelialized vasculature. Biomaterials 2015; 40:72-9. [DOI: 10.1016/j.biomaterials.2014.11.027] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 11/01/2014] [Accepted: 11/08/2014] [Indexed: 02/08/2023]
|
132
|
Caralt M, Uzarski JS, Iacob S, Obergfell KP, Berg N, Bijonowski BM, Kiefer KM, Ward HH, Wandinger-Ness A, Miller WM, Zhang ZJ, Abecassis MM, Wertheim JA. Optimization and critical evaluation of decellularization strategies to develop renal extracellular matrix scaffolds as biological templates for organ engineering and transplantation. Am J Transplant 2015; 15:64-75. [PMID: 25403742 PMCID: PMC4276475 DOI: 10.1111/ajt.12999] [Citation(s) in RCA: 165] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 08/14/2014] [Accepted: 08/30/2014] [Indexed: 01/25/2023]
Abstract
The ability to generate patient-specific cells through induced pluripotent stem cell (iPSC) technology has encouraged development of three-dimensional extracellular matrix (ECM) scaffolds as bioactive substrates for cell differentiation with the long-range goal of bioengineering organs for transplantation. Perfusion decellularization uses the vasculature to remove resident cells, leaving an intact ECM template wherein new cells grow; however, a rigorous evaluative framework assessing ECM structural and biochemical quality is lacking. To address this, we developed histologic scoring systems to quantify fundamental characteristics of decellularized rodent kidneys: ECM structure (tubules, vessels, glomeruli) and cell removal. We also assessed growth factor retention--indicating matrix biofunctionality. These scoring systems evaluated three strategies developed to decellularize kidneys (1% Triton X-100, 1% Triton X-100/0.1% sodium dodecyl sulfate (SDS) and 0.02% Trypsin-0.05% EGTA/1% Triton X-100). Triton and Triton/SDS preserved renal microarchitecture and retained matrix-bound basic fibroblast growth factor and vascular endothelial growth factor. Trypsin caused structural deterioration and growth factor loss. Triton/SDS-decellularized scaffolds maintained 3 h of leak-free blood flow in a rodent transplantation model and supported repopulation with human iPSC-derived endothelial cells and tubular epithelial cells ex vivo. Taken together, we identify an optimal Triton/SDS-based decellularization strategy that produces a biomatrix that may ultimately serve as a rodent model for kidney bioengineering.
Collapse
Affiliation(s)
- Mireia Caralt
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611,Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611,Servei Cirurgia HepatoBilioPancreatica i Trasplantaments. Hospital Universitari Vall Hebron. Universitat Autonoma de Barcelona. Spain
| | - Joseph S. Uzarski
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611,Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Stanca Iacob
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611,Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Kyle P. Obergfell
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
| | - Natasha Berg
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Brent M. Bijonowski
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611,Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Kathryn M. Kiefer
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
| | - Heather H. Ward
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM, 87131
| | | | - William M. Miller
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, 60201,Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60201
| | - Zheng J. Zhang
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611,Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Michael M. Abecassis
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611,Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Jason A. Wertheim
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611,Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611,Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60201,Department of Surgery, Jesse Brown VA Medical Center, Chicago, IL, 60612,Institute for BioNanotechnology in Medicine, Northwestern University, Chicago, IL, 60611,Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60201,Address for correspondence: Jason A. Wertheim, M.D., Ph.D., 676 St. Clair St. Suite 1900, Chicago, Illinois 60611, Telephone: (312) 695-0257, Fax: (312) 503-3366,
| |
Collapse
|
133
|
Michel SG, Madariaga MLL, Villani V, Shanmugarajah K. Current progress in xenotransplantation and organ bioengineering. Int J Surg 2014; 13:239-244. [PMID: 25496853 DOI: 10.1016/j.ijsu.2014.12.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 11/30/2014] [Accepted: 12/07/2014] [Indexed: 12/25/2022]
Abstract
Organ transplantation represents a unique method of treatment to cure people with end-stage organ failure. Since the first successful organ transplant in 1954, the field of transplantation has made great strides forward. However, despite the ability to transform and save lives, transplant surgery is still faced with a fundamental problem the number of people requiring organ transplants is simply higher than the number of organs available. To put this in stark perspective, because of this critical organ shortage 18 people every day in the United States alone die on a transplant waiting list (U.S. Department of Health & Human Services, http://organdonor.gov/about/data.html). To address this problem, attempts have been made to increase the organ supply through xenotransplantation and more recently, bioengineering. Here we trace the development of both fields, discuss their current status and highlight limitations going forward. Ultimately, lessons learned in each field may prove widely applicable and lead to the successful development of xenografts, bioengineered constructs, and bioengineered xeno-organs, thereby increasing the supply of organs for transplantation.
Collapse
Affiliation(s)
- Sebastian G Michel
- Transplantation Biology Research Center, Massachusetts General Hospital, Building 149, 13th Street, Charlestown, Boston, MA 02114, USA; Department of Cardiac Surgery, Ludwig-Maximilians-Universität München, Munich D-81377, Germany.
| | - Maria Lucia L Madariaga
- Transplantation Biology Research Center, Massachusetts General Hospital, Building 149, 13th Street, Charlestown, Boston, MA 02114, USA; Department of Surgery, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02129, USA
| | - Vincenzo Villani
- Transplantation Biology Research Center, Massachusetts General Hospital, Building 149, 13th Street, Charlestown, Boston, MA 02114, USA
| | - Kumaran Shanmugarajah
- Transplantation Biology Research Center, Massachusetts General Hospital, Building 149, 13th Street, Charlestown, Boston, MA 02114, USA; Division of Surgery, Imperial College London, Exhibition Road, London SW7 2AZ, United Kingdom.
| |
Collapse
|
134
|
Mitragotri S, Burke PA, Langer R. Overcoming the challenges in administering biopharmaceuticals: formulation and delivery strategies. Nat Rev Drug Discov 2014; 13:655-72. [PMID: 25103255 PMCID: PMC4455970 DOI: 10.1038/nrd4363] [Citation(s) in RCA: 1153] [Impact Index Per Article: 104.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The formulation and delivery of biopharmaceutical drugs, such as monoclonal antibodies and recombinant proteins, poses substantial challenges owing to their large size and susceptibility to degradation. In this Review we highlight recent advances in formulation and delivery strategies--such as the use of microsphere-based controlled-release technologies, protein modification methods that make use of polyethylene glycol and other polymers, and genetic manipulation of biopharmaceutical drugs--and discuss their advantages and limitations. We also highlight current and emerging delivery routes that provide an alternative to injection, including transdermal, oral and pulmonary delivery routes. In addition, the potential of targeted and intracellular protein delivery is discussed.
Collapse
Affiliation(s)
- Samir Mitragotri
- Department of Chemical Engineering, Center for Bioengineering, University of California, Santa Barbara, California 92106, USA
| | - Paul A Burke
- Burke Bioventures LLC, 277 Broadway, Cambridge, Massachusetts 02139, USA
| | - Robert Langer
- Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
135
|
Salvatori M, Peloso A, Katari R, Orlando G. Regeneration and bioengineering of the kidney: current status and future challenges. Curr Urol Rep 2014; 15:379. [PMID: 24375058 DOI: 10.1007/s11934-013-0379-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The prevalence of chronic kidney disease continues to outpace the development of effective treatment strategies. For patients with advanced disease, renal replacement therapies approximate the filtration functions of the kidney at considerable cost and inconvenience, while failing to restore the resorptive and endocrine functions. Allogeneic transplantation remains the only restorative treatment, but donor shortage, surgical morbidity and the need for lifelong immunosuppression significantly limit clinical application. Emerging technologies in the fields of regenerative medicine and tissue engineering strive to address these limitations. We review recent advances in cell-based therapies, primordial allografts, bio-artificial organs and whole-organ bioengineering as they apply to renal regeneration. Collaborative efforts across these fields aim to produce a bioengineered kidney capable of restoring renal function in patients with end-stage disease.
Collapse
|
136
|
Tissue engineered scaffolds for an effective healing and regeneration: reviewing orthotopic studies. BIOMED RESEARCH INTERNATIONAL 2014; 2014:398069. [PMID: 25250319 PMCID: PMC4163448 DOI: 10.1155/2014/398069] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/22/2014] [Indexed: 12/20/2022]
Abstract
It is commonly stated that tissue engineering is the most promising approach to treat or replace failing tissues/organs. For this aim, a specific strategy should be planned including proper selection of biomaterials, fabrication techniques, cell lines, and signaling cues. A great effort has been pursued to develop suitable scaffolds for the restoration of a variety of tissues and a huge number of protocols ranging from in vitro to in vivo studies, the latter further differentiating into several procedures depending on the type of implantation (i.e., subcutaneous or orthotopic) and the model adopted (i.e., animal or human), have been developed. All together, the published reports demonstrate that the proposed tissue engineering approaches spread toward multiple directions. The critical review of this scenario might suggest, at the same time, that a limited number of studies gave a real improvement to the field, especially referring to in vivo investigations. In this regard, the present paper aims to review the results of in vivo tissue engineering experimentations, focusing on the role of the scaffold and its specificity with respect to the tissue to be regenerated, in order to verify whether an extracellular matrix-like device, as usually stated, could promote an expected positive outcome.
Collapse
|
137
|
Zambon JP, Magalhaes RS, Ko I, Ross CL, Orlando G, Peloso A, Atala A, Yoo JJ. Kidney regeneration: Where we are and future perspectives. World J Nephrol 2014; 3:24-30. [PMID: 25332894 PMCID: PMC4202490 DOI: 10.5527/wjn.v3.i3.24] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 06/27/2014] [Accepted: 07/29/2014] [Indexed: 02/06/2023] Open
Abstract
In 2012, about 16487 people received kidney transplants in the United States, whereas 95022 candidates were on the waiting list by the end of the year. Despite advances in renal transplant immunology, approximately 40% of recipients will die or lose graft within 10 years. The limitations of current therapies for renal failure have led researchers to explore the development of modalities that could improve, restore, or replace the renal function. The aim of this paper is to describe a reasonable approach for kidney regeneration and review the current literature regarding cell sources and mechanisms to develop a bioengineering kidney. Due to kidneys peculiar anatomy, extracellular matrix based scaffolds are rational starting point for their regeneration. The perfusion of detergents through the kidney vasculature is an efficient method for delivering decellularizing agents to cells and for removing of cellular material from the tissue. Many efforts have focused on the search of a reliable cell source to provide enrichment for achieving stable renal cell systems. For an efficient bioengineered kidney, these cells must be attached to the organ and then maturated into the bioractors, which simulates the human body environment. A functional bioengineered kidney is still a big challenge for scientists. In the last ten years we have got many improvements on the field of solid organ regeneration; however, we are still far away from the main target. Currently, regenerative centers worldwide have been striving to find feasible strategies to develop bioengineered kidneys. Cell-scaffold technology gives hope to end-stage renal disease patients who struggle with morbidity and mortality due to extended periods on dialysis or immunosupression. The potential of bioengineered organ is to provide a reliable source of organs, which can be refunctionalized and transplanted.
Collapse
|
138
|
Schuessler TK, Chan XY, Chen HJ, Ji K, Park KM, Roshan-Ghias A, Sethi P, Thakur A, Tian X, Villasante A, Zervantonakis IK, Moore NM, Nagahara LA, Kuhn NZ. Biomimetic tissue-engineered systems for advancing cancer research: NCI Strategic Workshop report. Cancer Res 2014; 74:5359-63. [PMID: 25095784 DOI: 10.1158/0008-5472.can-14-1706] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Advanced technologies and biomaterials developed for tissue engineering and regenerative medicine present tractable biomimetic systems with potential applications for cancer research. Recently, the National Cancer Institute convened a Strategic Workshop to explore the use of tissue biomanufacturing for development of dynamic, physiologically relevant in vitro and ex vivo biomimetic systems to study cancer biology and drug efficacy. The workshop provided a forum to identify current progress, research gaps, and necessary steps to advance the field. Opportunities discussed included development of tumor biomimetic systems with an emphasis on reproducibility and validation of new biomimetic tumor models, as described in this report.
Collapse
Affiliation(s)
| | - Xin Yi Chan
- Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Physical Sciences Oncology Center, Johns Hopkins University, Baltimore, Maryland
| | | | - Kyungmin Ji
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, Michigan
| | - Kyung Min Park
- Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Physical Sciences Oncology Center, Johns Hopkins University, Baltimore, Maryland
| | - Alireza Roshan-Ghias
- Department of Biomedical Engineering, Laboratory for Stem Cells and Tissue Engineering, Columbia University, New York, New York
| | - Pallavi Sethi
- Department of Pharmaceutical Sciences, Cancer Nanotechnology Training Center, University of Kentucky College of Pharmacy, Lexington, Kentucky
| | - Archana Thakur
- Department of Oncology, Karmanos Cancer Institute at Wayne State University, Detroit, Michigan
| | - Xi Tian
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Aranzazu Villasante
- Department of Biomedical Engineering, Laboratory for Stem Cells and Tissue Engineering, Columbia University, New York, New York
| | | | - Nicole M Moore
- Division of Cancer Biology, National Cancer Institute, Rockville, Maryland
| | - Larry A Nagahara
- Division of Cancer Biology, National Cancer Institute, Rockville, Maryland
| | - Nastaran Z Kuhn
- Division of Cancer Biology, National Cancer Institute, Rockville, Maryland.
| |
Collapse
|
139
|
Choi SH, Chun SY, Chae SY, Kim JR, Oh SH, Chung SK, Lee JH, Song PH, Choi GS, Kim TH, Kwon TG. Development of a porcine renal extracellular matrix scaffold as a platform for kidney regeneration. J Biomed Mater Res A 2014; 103:1391-403. [DOI: 10.1002/jbm.a.35274] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 06/08/2014] [Accepted: 07/03/2014] [Indexed: 12/24/2022]
Affiliation(s)
- Seock Hwan Choi
- Department of Urology; School of Medicine; Kyungpook National University; Daegu Korea
| | - So Young Chun
- Joint Institute for Regenerative Medicine; Kyungpook National University Hospital; Daegu Korea
| | - Seon Yeong Chae
- Joint Institute for Regenerative Medicine; Kyungpook National University Hospital; Daegu Korea
| | - Jin Rae Kim
- Department of Advanced Materials; Hannam University; Daejeon Korea
| | - Se Heang Oh
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine; Dankook University; Chungnam Korea
| | - Sung Kwang Chung
- Department of Urology; School of Medicine; Kyungpook National University; Daegu Korea
| | - Jin Ho Lee
- Department of Advanced Materials; Hannam University; Daejeon Korea
| | - Phil Hyun Song
- Department of Urology; College of Medicine, Yeungnam University; Daegu Korea
| | - Gyu-Seog Choi
- Department of Colorectal Cancer Center; School of Medicine; Kyungpook National University; Daegu Korea
| | - Tae-Hwan Kim
- Department of Urology; School of Medicine; Kyungpook National University; Daegu Korea
| | - Tae Gyun Kwon
- Department of Urology; School of Medicine; Kyungpook National University; Daegu Korea
- Joint Institute for Regenerative Medicine; Kyungpook National University Hospital; Daegu Korea
| |
Collapse
|
140
|
Li PS, Lee IL, Yu WL, Sun JS, Jane WN, Shen HH. A novel albumin-based tissue scaffold for autogenic tissue engineering applications. Sci Rep 2014; 4:5600. [PMID: 25034369 PMCID: PMC4102902 DOI: 10.1038/srep05600] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 06/19/2014] [Indexed: 01/12/2023] Open
Abstract
Tissue scaffolds provide a framework for living tissue regeneration. However, traditional tissue scaffolds are exogenous, composed of metals, ceramics, polymers, and animal tissues, and have a defined biocompatibility and application. This study presents a new method for obtaining a tissue scaffold from blood albumin, the major protein in mammalian blood. Human, bovine, and porcine albumin was polymerised into albumin polymers by microbial transglutaminase and was then cast by freeze-drying-based moulding to form albumin tissue scaffolds. Scanning electron microscopy and material testing analyses revealed that the albumin tissue scaffold possesses an extremely porous structure, moderate mechanical strength, and resilience. Using a culture of human mesenchymal stem cells (MSCs) as a model, we showed that MSCs can be seeded and grown in the albumin tissue scaffold. Furthermore, the albumin tissue scaffold can support the long-term osteogenic differentiation of MSCs. These results show that the albumin tissue scaffold exhibits favourable material properties and good compatibility with cells. We propose that this novel tissue scaffold can satisfy essential needs in tissue engineering as a general-purpose substrate. The use of this scaffold could lead to the development of new methods of artificial fabrication of autogenic tissue substitutes.
Collapse
Affiliation(s)
- Pei-Shan Li
- 1] Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan [2] Tissue Regeneration Product Technology Division, Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu County 310, Taiwan [3]
| | - I-Liang Lee
- 1] c/o Rm. 624, Bldg. 53, No. 195, Sec. 4, Chung Hsing Rd., Chutung Township, Hsinchu County 310, Taiwan [2]
| | - Wei-Lin Yu
- Tissue Regeneration Product Technology Division, Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu County 310, Taiwan
| | - Jui-Sheng Sun
- Department of Orthopedic Surgery, School of Medicine, College of Medicine, National Taiwan University, Taipei 10051 and Department of Orthopaedic Surgery, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu 30059, Taiwan
| | - Wann-Neng Jane
- Institute of Plant and Microbial Biology, Academia Sinica, Nankang, Taipei 11529, Taiwan
| | - Hsin-Hsin Shen
- Tissue Regeneration Product Technology Division, Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu County 310, Taiwan
| |
Collapse
|
141
|
Abstract
One in 10 Americans suffers from chronic kidney disease, and close to 90,000 people die each year from causes related to kidney failure. Patients with end-stage renal disease are faced with two options: hemodialysis or transplantation. Unfortunately, the transplantation option is limited because of the shortage of donor organs and the need for immunosuppression. Bioengineered kidney grafts theoretically present a novel solution to both problems. Herein, we discuss the history of bioengineering organs, the current status of bioengineered kidneys, considerations for the future of the field, and challenges to clinical translation. We hope that by integrating principles of tissue engineering, and stem cell and developmental biology, bioengineered kidney grafts will advance the field of regenerative medicine while meeting a critical clinical need.
Collapse
Affiliation(s)
- Maria Lucia L Madariaga
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA; Harvard Medical School, Harvard Stem Cell Institute, Boston, MA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Harald C Ott
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA; Harvard Medical School, Harvard Stem Cell Institute, Boston, MA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA.
| |
Collapse
|
142
|
Guyette JP, Gilpin SE, Charest JM, Tapias LF, Ren X, Ott HC. Perfusion decellularization of whole organs. Nat Protoc 2014; 9:1451-68. [PMID: 24874812 DOI: 10.1038/nprot.2014.097] [Citation(s) in RCA: 176] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The native extracellular matrix (ECM) outlines the architecture of organs and tissues. It provides a unique niche of composition and form, which serves as a foundational scaffold that supports organ-specific cell types and enables normal organ function. Here we describe a standard process for pressure-controlled perfusion decellularization of whole organs for generating acellular 3D scaffolds with preserved ECM protein content, architecture and perfusable vascular conduits. By applying antegrade perfusion of detergents and subsequent washes to arterial vasculature at low physiological pressures, successful decellularization of complex organs (i.e., hearts, lungs and kidneys) can be performed. By using appropriate modifications, pressure-controlled perfusion decellularization can be achieved in small-animal experimental models (rat organs, 4-5 d) and scaled to clinically relevant models (porcine and human organs, 12-14 d). Combining the unique structural and biochemical properties of native acellular scaffolds with subsequent recellularization techniques offers a novel platform for organ engineering and regeneration, for experimentation ex vivo and potential clinical application in vivo.
Collapse
Affiliation(s)
- Jacques P Guyette
- 1] Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA. [2] Harvard Medical School, Boston, Massachusetts, USA. [3]
| | - Sarah E Gilpin
- 1] Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA. [2] Harvard Medical School, Boston, Massachusetts, USA. [3]
| | - Jonathan M Charest
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Luis F Tapias
- 1] Harvard Medical School, Boston, Massachusetts, USA. [2] Department of Surgery, Division of Thoracic Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Xi Ren
- 1] Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA. [2] Harvard Medical School, Boston, Massachusetts, USA
| | - Harald C Ott
- 1] Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA. [2] Harvard Medical School, Boston, Massachusetts, USA. [3] Department of Surgery, Division of Thoracic Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA. [4] Harvard Stem Cell Institute, Boston, Massachusetts, USA
| |
Collapse
|
143
|
Yu YL, Shao YK, Ding YQ, Lin KZ, Chen B, Zhang HZ, Zhao LN, Wang ZB, Zhang JS, Tang ML, Mei J. Decellularized kidney scaffold-mediated renal regeneration. Biomaterials 2014; 35:6822-8. [PMID: 24855960 DOI: 10.1016/j.biomaterials.2014.04.074] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 04/22/2014] [Indexed: 01/04/2023]
Abstract
Renal regeneration approaches offer great potential for the treatment of chronic kidney disease, but their availability remains limited by the clinical challenges they pose. In the present study, we used continuous detergent perfusion to generate decellularized (DC) rat kidney scaffolds. The scaffolds retained intact vascular trees and overall architecture, along with significant concentrations of various cytokines, but lost all cellular components. To evaluate its potential in renal function recovery, DC scaffold tissue was grafted onto partially nephrectomized rat kidneys. An increase of renal size was found, and regenerated renal parenchyma cells were observed in the repair area containing the grafted scaffold. In addition, the number of nestin-positive renal progenitor cells was markedly higher in scaffold-grafted kidneys compared to controls. Moreover, radionuclide scan analysis showed significant recovery of renal functions at 6 weeks post-implantation. Our results provide further evidence to show that DC kidney scaffolds could be used to promote renal recovery in the treatment of chronic kidney disease.
Collapse
Affiliation(s)
- Y L Yu
- Anatomy Department, Wenzhou Medical University, Wenzhou 325035, China; Institute of Bioscaffold Transplantation and Immunology, Wenzhou Medical University, Wenzhou 325035, China
| | - Y K Shao
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Y Q Ding
- Institute of Neuroscience, Wenzhou Medical University, Wenzhou 325035, China
| | - K Z Lin
- Institute of Bioscaffold Transplantation and Immunology, Wenzhou Medical University, Wenzhou 325035, China
| | - B Chen
- Institute of Bioscaffold Transplantation and Immunology, Wenzhou Medical University, Wenzhou 325035, China; Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 32000, China
| | - H Z Zhang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 32000, China
| | - L N Zhao
- Anatomy Department, Wenzhou Medical University, Wenzhou 325035, China; Institute of Bioscaffold Transplantation and Immunology, Wenzhou Medical University, Wenzhou 325035, China
| | - Z B Wang
- Institute of Bioscaffold Transplantation and Immunology, Wenzhou Medical University, Wenzhou 325035, China
| | - J S Zhang
- Anatomy Department, Wenzhou Medical University, Wenzhou 325035, China; Institute of Bioscaffold Transplantation and Immunology, Wenzhou Medical University, Wenzhou 325035, China
| | - M L Tang
- Anatomy Department, Wenzhou Medical University, Wenzhou 325035, China; Institute of Bioscaffold Transplantation and Immunology, Wenzhou Medical University, Wenzhou 325035, China
| | - J Mei
- Anatomy Department, Wenzhou Medical University, Wenzhou 325035, China; Institute of Bioscaffold Transplantation and Immunology, Wenzhou Medical University, Wenzhou 325035, China; Institute of Neuroscience, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
144
|
Abstract
BACKGROUND Kidney regeneration is currently gaining considerable attention in place of kidney dialysis as the ultimate therapeutic strategy for renal failure. However, because of anatomical complications, the kidney is believed to be the hardest organ to regenerate. Such a complicated organ is virtually impossible to imagine being completely rebuilt de novo from stem cells. Nevertheless, several research groups are attempting this large challenge. SUMMARY There are 4 major strategies for de novo kidney regeneration from stem cells. These strategies include the use of: (i) a decellularized cadaveric scaffold, (ii) blastocyst decomplementation, (iii) a nephrogenic niche for growing a xeno-embyro, and (iv) self-assembly potential. All of these strategies may be applicable in the clinical setting, but a substantial preparation period appears to be required. KEY MESSAGES Although many outstanding problems remain for kidney regeneration, including ethical issues and the formation of chimeric structures, trials provide hope for dialysis patients and kidney regeneration is expected to be a reality in the future.
Collapse
Affiliation(s)
- Takashi Yokoo
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
145
|
Katari R, Peloso A, Zambon JP, Soker S, Stratta RJ, Atala A, Orlando G. Renal bioengineering with scaffolds generated from human kidneys. Nephron Clin Pract 2014; 126:119. [PMID: 24854653 DOI: 10.1159/000360684] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND In 2012, about 16,487 people received kidney transplants in the USA whereas 95,022 candidates were on the waiting list at the end of the year. Moreover, more than 2,600 kidneys procured annually for transplantation are discarded for a variety of reasons. We hypothesize that this pool of discarded kidneys could in part meet the growing, urgent need for transplantable kidneys using current methods for organ bioengineering and regeneration and surgical transplantation. The recellularization of extracellular matrix (ECM) scaffolds has the potential to meet the uniquely ambitious engineering challenges posed by complex solid organs such as the kidney. SUMMARY Attempts to manufacture and implant simpler, hollow structures such as bladders, vessels, urethras, and segments of the upper airways have been successful in the short and mid terms. However, the bioengineering of complex solid organs such as the kidney is a more challenging task that requires a different approach. In previous studies, we showed that decellularized porcine kidneys yield renal ECM scaffolds that preserve their basic architecture and structural components, support cell growth in vivo and in vitro, and maintain a patent vasculature capable of sustaining physiological blood pressure. In a subsequent report, using the same methods, we found that detergent-based decellularization of discarded human renal kidneys preserved their innate ECM framework, biochemical properties, and angiogenic capacity and - importantly - a patent vascular network. Furthermore, the process resulted in the clearance of immunogenic antigens, which has monumental implications for clinical outcomes in the long term in terms of graft rejection. Consequently, these kidneys show promise in bioengineering and transplantation. We refer to this avenue of research and development as 'cell-scaffold technology'. KEY MESSAGES In 2011, more than 4,700 patients died while on the waiting list for a kidney transplant. In this context, we believe that cell-scaffold technology has the potential to form a bridge between regenerative medicine and transplantation surgery. These methods, in theory, could provide a potentially inexhaustible source of transplantable organs. Unfortunately, current investigations are still in their very early stages and clinical translation is not immediately available in the short term. Thus, identifying the most important obstacles confronting cell-scaffold technology and focusing research efforts in this direction will be important for advancing the state of the art and meeting the clinical needs. We believe that cell-scaffold technology research and development would benefit greatly from a deeper understanding of the physiological mechanisms underlying the natural organogenesis, regeneration, and repair that characterize embryonic humans and simpler organisms. Furthermore, the importance of vascularization - the fundamental caveat of modern surgery - cannot be overstated, especially when discussing the implantation of de novo organs.
Collapse
Affiliation(s)
- Ravi Katari
- Wake Forest School of Medicine, Winston-Salem, N.C., USA
| | | | | | | | | | | | | |
Collapse
|
146
|
Abstract
PURPOSE OF REVIEW Patients suffering from end-stage organ failure requiring organ transplantation face donor organ shortage and adverse effect of chronic immunosuppression. Recent progress in the field of organ bioengineering based on decellularized organ scaffolds and patient-derived cells holds great promise to address these issues. RECENT FINDINGS Perfusion-decellularization is the most consistent method to obtain decellularized whole-organ scaffolds to serve as a platform for organ bioengineering. Important advances have occurred in organ bioengineering using decellularized scaffolds in small animal models. However, the function exhibited by bioengineered organs has been rudimentary. Pluripotent stem cells seem to hold promise as the ideal regenerative cells to be used with this approach but the techniques to effectively and reliably manipulate their fate are still to be discovered. Finally, this technology needs to be scaled up to human size to be of clinical relevance. SUMMARY The search for alternatives to allogeneic organ transplantation continues. Important milestones have been achieved in organ bioengineering with the use of decellularized scaffolds. However, many challenges remain on the way to producing an autologous, fully functional organ that can be transplanted similar to a donor organ.
Collapse
|
147
|
Calle EA, Ghaedi M, Sundaram S, Sivarapatna A, Tseng MK, Niklason LE. Strategies for whole lung tissue engineering. IEEE Trans Biomed Eng 2014; 61:1482-96. [PMID: 24691527 PMCID: PMC4126648 DOI: 10.1109/tbme.2014.2314261] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Recent work has demonstrated the feasibility of using decellularized lung extracellular matrix scaffolds to support the engineering of functional lung tissue in vitro. Rendered acellular through the use of detergents and other reagents, the scaffolds are mounted in organ-specific bioreactors where cells in the scaffold are provided with nutrients and appropriate mechanical stimuli such as ventilation and perfusion. Though initial studies are encouraging, a great deal remains to be done to advance the field and transition from rodent lungs to whole human tissue engineered lungs. To do so, a variety of hurdles must be overcome. In particular, a reliable source of human-sized scaffolds, as well as a method of terminal sterilization of scaffolds, must be identified. Continued research in lung cell and developmental biology will hopefully help identify the number and types of cells that will be required to regenerate functional lung tissue. Finally, bioreactor designs must be improved in order to provide more precise ventilation stimuli and vascular perfusion in order to avoid injury to or death of the cells cultivated within the scaffold. Ultimately, the success of efforts to engineer a functional lung in vitro will critically depend on the ability to create a fully endothelialized vascular network that provides sufficient barrier function and alveolar-capillary surface area to exchange gas at rates compatible with healthy lung function.
Collapse
Affiliation(s)
- Elizabeth A. Calle
- Department of Biomedical Engineering, Yale University, New Haven, CT 06519 USA
| | - Mahboobe Ghaedi
- Department of Anesthesia, Yale University, New Haven, CT 06519 USA
| | - Sumati Sundaram
- Department of Anesthesia, Yale University, New Haven, CT 06519 USA
| | - Amogh Sivarapatna
- Department of Biomedical Engineering, Yale University, New Haven, CT 06519 USA
| | - Michelle K. Tseng
- Department of Biomedical Engineering, Yale University, New Haven, CT 06519 USA
| | - Laura E. Niklason
- Department of Anesthesia and Department of Biomedical Engineering, Yale University, New Haven, CT 06519 USA
| |
Collapse
|
148
|
Nowacki M, Kloskowski T, Pokrywczyńska M, Nazarewski Ł, Jundziłł A, Pietkun K, Tyloch D, Rasmus M, Warda K, Habib SL, Drewa T. Is regenerative medicine a new hope for kidney replacement? J Artif Organs 2014; 17:123-34. [DOI: 10.1007/s10047-014-0767-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 04/01/2014] [Indexed: 12/24/2022]
|
149
|
DesRochers TM, Palma E, Kaplan DL. Tissue-engineered kidney disease models. Adv Drug Deliv Rev 2014; 69-70:67-80. [PMID: 24361391 DOI: 10.1016/j.addr.2013.12.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 12/08/2013] [Accepted: 12/09/2013] [Indexed: 02/08/2023]
Abstract
Renal disease represents a major health problem that often results in end-stage renal failure necessitating dialysis and eventually transplantation. Historically these diseases have been studied with patient observation and screening, animal models, and two-dimensional cell culture. In this review, we focus on recent advances in tissue engineered kidney disease models that have the capacity to compensate for the limitations of traditional modalities. The cells and materials utilized to develop these models are discussed and tissue engineered models of polycystic kidney disease, drug-induced nephrotoxicity, and the glomerulus are examined in detail. The application of these models has the potential to direct future disease treatments and preclinical drug development.
Collapse
|
150
|
Moran EC, Dhal A, Vyas D, Lanas A, Soker S, Baptista PM. Whole-organ bioengineering: current tales of modern alchemy. Transl Res 2014; 163:259-67. [PMID: 24486135 DOI: 10.1016/j.trsl.2014.01.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 01/06/2014] [Indexed: 12/31/2022]
Abstract
End-stage organ disease affects millions of people around the world, to whom organ transplantation is the only definitive cure available. However, persistent organ shortage and the resulting widespread transplant backlog are part of a disturbing reality and a common burden felt by thousands of patients on waiting lists in almost every country where organ transplants are performed. Several alternatives and potential solutions to this problem have been sought in past decades, but one seems particularly promising now: whole-organ bioengineering. This review describes briefly the evolution of organ transplantation and the development of decellularized organ scaffolds and their application to organ bioengineering. This modern alchemy of generating whole-organ scaffolds and recellularizing them with multiple cell types in perfusion bioreactors is paving the way for a new revolution in transplantation medicine. Furthermore, although the first generation of bioengineered organs still lacks true clinical value, it has created a number of novel tissue and organ model platforms with direct application in other areas of science (eg, developmental biology and stem cell biology, drug discovery, physiology and metabolism). In this review, we describe the current status and numerous applications of whole-organ bioengineering, focusing also on the multiple challenges that researchers have to overcome to translate these novel technologies fully into transplantation medicine.
Collapse
Affiliation(s)
- Emma C Moran
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC
| | - Abritee Dhal
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC
| | - Dipen Vyas
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC
| | - Angel Lanas
- University of Zaragoza, Zaragoza, Spain; IIS Aragón, CIBERehd, Zaragoza, Spain
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC
| | - Pedro M Baptista
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC; Aragon Health Sciences Institute, Zaragoza, Spain.
| |
Collapse
|