1
|
Nazemidashtarjandi S, Larsen B, Cheng K, Faulkner S, Peppas NA, Parekh SH, Zoldan J. Near-infrared light-responsive hydrogels for on-demand dual delivery of proangiogenic growth factors. Acta Biomater 2024; 183:61-73. [PMID: 38838911 PMCID: PMC11514431 DOI: 10.1016/j.actbio.2024.05.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024]
Abstract
Achieving precise spatiotemporal control over the release of proangiogenic factors is crucial for vasculogenesis, the process of de novo blood vessel formation. Although various strategies have been explored, there is still a need to develop cell-laden biomaterials with finely controlled release of proangiogenic factors at specific locations and time points. We report on the developed of a near-infrared (NIR) light-responsive collagen hydrogel comprised of gold nanorods (GNRs)-conjugated liposomes containing proangiogenic growth factors (GFs). We demonstrated that this system enables on-demand dual delivery of GFs at specific sites and over selected time intervals. Liposomes were strategically formulated to encapsulate either platelet-derived growth factor (PDGF) or vascular endothelial growth factor (VEGF), each conjugated to gold nanorods (GNRs) with distinct geometries and surface plasmon resonances at 710 nm (GNR710) and 1064 nm (GNR1064), respectively. Using near infrared (NIR) irradiation and two-photon (2P) luminescence imaging, we successfully demonstrated the independent release of PDGF from GNR710 conjugated liposomes and VEGF from GNR1064-conjugated liposomes. Our imaging data revealed rapid release kinetics, with localized PDGF released in approximately 4 min and VEGF in just 1 and a half minutes following NIR laser irradiation. Importantly, we demonstrated that the release of each GF could be independently triggered using NIR irradiation with the other GF formulation remaining retained within the liposomes. This light-responsive collagen hydrogels holds promise for various applications in regenerative medicine where the establishment of a guided vascular network is essential for the survival and integration of engineered tissues. STATEMENT OF SIGNIFICANCE: In this study, we have developed a light-responsive system with gold nanorods (GNRs)-conjugated liposomes in a collagen hydrogel, enabling precise dual delivery of proangiogenic growth factors (GFs) at specific locations and timepoints. Liposomes, containing platelet-derived growth factor (PDGF) or vascular endothelial growth factor (VEGF), release independently under near- infrared irradiation. This approach allows external activation of desired GF release, ensuring high cell viability. Each GF can be triggered independently, retaining the other within the liposomes. Beyond its application in establishing functional vascular networks, this dual delivery system holds promise as a universal platform for delivering various combinations of two or more GFs.
Collapse
Affiliation(s)
- Saeed Nazemidashtarjandi
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78711, United Sates
| | - Bryce Larsen
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78711, United Sates
| | - Kristie Cheng
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78711, United Sates
| | - Sara Faulkner
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78711, United Sates
| | - Nicholas A Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78711, United Sates; Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78711, United Sates
| | - Sapun H Parekh
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78711, United Sates
| | - Janet Zoldan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78711, United Sates.
| |
Collapse
|
2
|
Jeon S, Lee YS, Oh SR, Jeong J, Lee DH, So KH, Hwang NS. Recent advances in endocrine organoids for therapeutic application. Adv Drug Deliv Rev 2023; 199:114959. [PMID: 37301512 DOI: 10.1016/j.addr.2023.114959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/21/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023]
Abstract
The endocrine system, consisting of the hypothalamus, pituitary, endocrine glands, and hormones, plays a critical role in hormone metabolic interactions. The complexity of the endocrine system is a significant obstacle to understanding and treating endocrine disorders. Notably, advances in endocrine organoid generation allow a deeper understanding of the endocrine system by providing better comprehension of molecular mechanisms of pathogenesis. Here, we highlight recent advances in endocrine organoids for a wide range of therapeutic applications, from cell transplantation therapy to drug toxicity screening, combined with development in stem cell differentiation and gene editing technologies. In particular, we provide insights into the transplantation of endocrine organoids to reverse endocrine dysfunctions and progress in developing strategies for better engraftments. We also discuss the gap between preclinical and clinical research. Finally, we provide future perspectives for research on endocrine organoids for the development of more effective treatments for endocrine disorders.
Collapse
Affiliation(s)
- Suwan Jeon
- Interdisciplinary Program for Biochemical Engineering and Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Young-Sun Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Seh Ri Oh
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Jinseong Jeong
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Dong-Hyun Lee
- Interdisciplinary Program for Biochemical Engineering and Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyoung-Ha So
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea; Bio-MAX/N-Bio Institute, Institute of Bio-Engineering, Seoul National University, Seoul 08826, Republic of Korea.
| | - Nathaniel S Hwang
- Interdisciplinary Program for Biochemical Engineering and Biotechnology, Seoul National University, Seoul 08826, Republic of Korea; School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea; Bio-MAX/N-Bio Institute, Institute of Bio-Engineering, Seoul National University, Seoul 08826, Republic of Korea; Institute of Engineering Research, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
3
|
Voznesenskaya A, Berggren PO, Ilegems E. Sustained heterologous gene expression in pancreatic islet organoids using adeno-associated virus serotype 8. Front Bioeng Biotechnol 2023; 11:1147244. [PMID: 37545890 PMCID: PMC10400289 DOI: 10.3389/fbioe.2023.1147244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 07/06/2023] [Indexed: 08/08/2023] Open
Abstract
Genetic modification of pancreatic islet organoids, assembled in vitro prior to transplantation is an emerging alternative to direct in vivo genetic manipulations for a number of clinical and research applications. We have previously shown that dispersion of islet cells followed by re-aggregation into islet organoids, or pseudoislets, allows for efficient transduction with viral vectors, while maintaining physiological functions of native islets. Among viruses currently used for genetic manipulations, adeno-associated viruses (AAVs) have the most attractive safety profile making them suitable for gene therapy applications. Studies reporting on pseudoislet transduction with AAVs are, however, lacking. Here, we have characterized in detail the performance of AAV serotype 8 in transduction of islet cells during pseudoislet formation in comparison with human adenovirus type 5 (AdV5). We have assessed such parameters as transduction efficiency, expression kinetics, and endocrine cell tropism of AAV8 alone or in combination with AdV5. Data provided within our study may serve as a reference point for future functional studies using AAVs for gene transfer to islet cell organoids and will facilitate further development of engineered pseudoislets of superior quality suitable for clinical transplantation.
Collapse
|
4
|
Wang Z, Jiang Z, Lu R, Kou L, Zhao YZ, Yao Q. Formulation strategies to provide oxygen-release to contrast local hypoxia for transplanted islets. Eur J Pharm Biopharm 2023; 187:130-140. [PMID: 37105362 DOI: 10.1016/j.ejpb.2023.04.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/08/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023]
Abstract
Islet transplantation refers to the transfusion of healthy islet cells into the diabetic recipients and reconstruction of their endogenous insulin secretion to achieve insulin independence. It is a minimally invasive surgery that holds renewed prospect as a therapeutic method for type 1 diabetes mellitus. However, poor oxygenation in the early post-transplantation period is considered as one of the major causes of islet loss and dysfunction. Due to the metabolism chacteristics, islets required a high supply of oxygen for cell survival while a hypoxia environment would lead to severe islet loss and graft failure. Emerging strategies have been proposed, including providing external oxygen and speeding up revascularization. From the perspective of formulation science, it is feasible and practical to protect transplanted islets by oxygen-release before revascularization as opposed to local hypoxia. In this study, we review the potential formulation strategies that could provide oxygen-release by either delivering external oxygen or triggering localized oxygen generation for transplanted islets.
Collapse
Affiliation(s)
- Zeqing Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Zhikai Jiang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Ruijie Lu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Longfa Kou
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Ying-Zheng Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Qing Yao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China.
| |
Collapse
|
5
|
Zeynaloo E, Stone LD, Dikici E, Ricordi C, Deo SK, Bachas LG, Daunert S, Lanzoni G. Delivery of therapeutic agents and cells to pancreatic islets: Towards a new era in the treatment of diabetes. Mol Aspects Med 2022; 83:101063. [PMID: 34961627 PMCID: PMC11328325 DOI: 10.1016/j.mam.2021.101063] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 02/07/2023]
Abstract
Pancreatic islet cells, and in particular insulin-producing beta cells, are centrally involved in the pathogenesis of diabetes mellitus. These cells are of paramount importance for the endocrine control of glycemia and glucose metabolism. In Type 1 Diabetes, islet beta cells are lost due to an autoimmune attack. In Type 2 Diabetes, beta cells become dysfunctional and insufficient to counterbalance insulin resistance in peripheral tissues. Therapeutic agents have been developed to support the function of islet cells, as well as to inhibit deleterious immune responses and inflammation. Most of these agents have undesired effects due to systemic administration and off-target effects. Typically, only a small fraction of therapeutic agent reaches the desired niche in the pancreas. Because islets and their beta cells are scattered throughout the pancreas, access to the niche is limited. Targeted delivery to pancreatic islets could dramatically improve the therapeutic effect, lower the dose requirements, and lower the side effects of agents administered systemically. Targeted delivery is especially relevant for those therapeutics for which the manufacturing is difficult and costly, such as cells, exosomes, and microvesicles. Along with therapeutic agents, imaging reagents intended to quantify the beta cell mass could benefit from targeted delivery. Several methods have been developed to improve the delivery of agents to pancreatic islets. Intra-arterial administration in the pancreatic artery is a promising surgical approach, but it has inherent risks. Targeted delivery strategies have been developed based on ligands for cell surface molecules specific to islet cells or inflamed vascular endothelial cells. Delivery methods range from nanocarriers and vectors to deliver pharmacological agents to viral and non-viral vectors for the delivery of genetic constructs. Several strategies demonstrated enhanced therapeutic effects in diabetes with lower amounts of therapeutic agents and lower off-target side effects. Microvesicles, exosomes, polymer-based vectors, and nanocarriers are gaining popularity for targeted delivery. Notably, liposomes, lipid-assisted nanocarriers, and cationic polymers can be bioengineered to be immune-evasive, and their advantages to transport cargos into target cells make them appealing for pancreatic islet-targeted delivery. Viral vectors have become prominent tools for targeted gene delivery. In this review, we discuss the latest strategies for targeted delivery of therapeutic agents and imaging reagents to pancreatic islet cells.
Collapse
Affiliation(s)
- Elnaz Zeynaloo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Chemistry, University of Miami, FL, USA.
| | - Logan D Stone
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Emre Dikici
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute - BioNIUM at University of Miami, Miami, FL, USA
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sapna K Deo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute - BioNIUM at University of Miami, Miami, FL, USA
| | - Leonidas G Bachas
- Department of Chemistry, University of Miami, FL, USA; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute - BioNIUM at University of Miami, Miami, FL, USA
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute - BioNIUM at University of Miami, Miami, FL, USA; Clinical and Translational Science Institute, University of Miami, Miami, FL, USA
| | - Giacomo Lanzoni
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA; Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute - BioNIUM at University of Miami, Miami, FL, USA.
| |
Collapse
|
6
|
Opara A, Jost A, Dagogo-Jack S, Opara EC. Islet cell encapsulation - Application in diabetes treatment. Exp Biol Med (Maywood) 2021; 246:2570-2578. [PMID: 34666516 PMCID: PMC8669170 DOI: 10.1177/15353702211040503] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In this minireview, we briefly outline the hallmarks of diabetes, the distinction between type 1 and type 2 diabetes, the global incidence of diabetes, and its associated comorbidities. The main goal of the review is to highlight the great potential of encapsulated pancreatic islet transplantation to provide a cure for type 1 diabetes. Following a short overview of the different approaches to islet encapsulation, we provide a summary of the merits and demerits of each approach of the encapsulation technology. We then discuss various attempts to clinical translation with each model of encapsulation as well as the factors that have mitigated the full clinical realization of the promise of the encapsulation technology, the progress that has been made and the challenges that remain to be overcome. In particular, we pay significant attention to the emerging strategies to overcome these challenges. We believe that these strategies to enhance the performance of the encapsulated islet constructs discussed herein provide good platforms for additional work to achieve successful clinical translation of the encapsulated islet technology.
Collapse
Affiliation(s)
- Amoge Opara
- Diabetes Section, Biologics Delivery Technologies, Reno, NV 89502, USA
| | - Alec Jost
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Sam Dagogo-Jack
- Division of Endocrinology, Diabetes & Metabolism, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Emmanuel C Opara
- Diabetes Section, Biologics Delivery Technologies, Reno, NV 89502, USA
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences (SBES), Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
7
|
Shin Low S, Nong Lim C, Yew M, Siong Chai W, Low LE, Manickam S, Ti Tey B, Show PL. Recent ultrasound advancements for the manipulation of nanobiomaterials and nanoformulations for drug delivery. ULTRASONICS SONOCHEMISTRY 2021; 80:105805. [PMID: 34706321 PMCID: PMC8555278 DOI: 10.1016/j.ultsonch.2021.105805] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/08/2021] [Accepted: 10/20/2021] [Indexed: 05/04/2023]
Abstract
Recent advances in ultrasound (US) have shown its great potential in biomedical applications as diagnostic and therapeutic tools. The coupling of US-assisted drug delivery systems with nanobiomaterials possessing tailor-made functions has been shown to remove the limitations of conventional drug delivery systems. The low-frequency US has significantly enhanced the targeted drug delivery effect and efficacy, reducing limitations posed by conventional treatments such as a limited therapeutic window. The acoustic cavitation effect induced by the US-mediated microbubbles (MBs) has been reported to replace drugs in certain acute diseases such as ischemic stroke. This review briefly discusses the US principles, with particular attention to the recent advancements in drug delivery applications. Furthermore, US-assisted drug delivery coupled with nanobiomaterials to treat different diseases (cancer, neurodegenerative disease, diabetes, thrombosis, and COVID-19) are discussed in detail. Finally, this review covers the future perspectives and challenges on the applications of US-mediated nanobiomaterials.
Collapse
Affiliation(s)
- Sze Shin Low
- Continental-NTU Corporate Lab, Nanyang Technological University, 50 Nanyang Drive, Singapore 637553, Singapore; Department of Chemical and Environmental Engineering, Faculty of Science and Engineering, University of Nottingham Malaysia, Jalan Broga, Semenyih 43500, Selangor Darul Ehsan, Malaysia
| | - Chang Nong Lim
- School of Engineering and Physical Sciences, Heriot-Watt University Malaysia, No. 1, Jalan Venna P5/2, Precinct 5, Putrajaya 62200, Malaysia
| | - Maxine Yew
- Department of Mechanical, Materials and Manufacturing Engineering, University of Nottingham Ningbo China, 199 Taikang East Road, Ningbo 315100, Zhejiang, China
| | - Wai Siong Chai
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, Guangdong, China
| | - Liang Ee Low
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia; Advanced Engineering Platform, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia; Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China.
| | - Sivakumar Manickam
- Petroleum and Chemical Engineering, Faculty of Engineering, Universiti Teknologi Brunei, Jalan Tungku Link Gadong, Bandar Seri Begawan, BE1410, Brunei Darussalam.
| | - Beng Ti Tey
- Advanced Engineering Platform, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia; Chemical Engineering Discipline, School of Engineering, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
| | - Pau Loke Show
- Department of Chemical and Environmental Engineering, Faculty of Science and Engineering, University of Nottingham Malaysia, Jalan Broga, Semenyih 43500, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
8
|
Alwahsh SM, Qutachi O, Starkey Lewis PJ, Bond A, Noble J, Burgoyne P, Morton N, Carter R, Mann J, Ferreira‐Gonzalez S, Alvarez‐Paino M, Forbes SJ, Shakesheff KM, Forbes S. Fibroblast growth factor 7 releasing particles enhance islet engraftment and improve metabolic control following islet transplantation in mice with diabetes. Am J Transplant 2021; 21:2950-2963. [PMID: 33428803 PMCID: PMC8603932 DOI: 10.1111/ajt.16488] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 12/20/2020] [Accepted: 01/05/2021] [Indexed: 01/25/2023]
Abstract
Transplantation of islets in type 1 diabetes (T1D) is limited by poor islet engraftment into the liver, with two to three donor pancreases required per recipient. We aimed to condition the liver to enhance islet engraftment to improve long-term graft function. Diabetic mice received a non-curative islet transplant (n = 400 islets) via the hepatic portal vein (HPV) with fibroblast growth factor 7-loaded galactosylated poly(DL-lactide-co-glycolic acid) (FGF7-GAL-PLGA) particles; 26-µm diameter particles specifically targeted the liver, promoting hepatocyte proliferation in short-term experiments: in mice receiving 0.1-mg FGF7-GAL-PLGA particles (60-ng FGF7) vs vehicle, cell proliferation was induced specifically in the liver with greater efficacy and specificity than subcutaneous FGF7 (1.25 mg/kg ×2 doses; ~75-µg FGF7). Numbers of engrafted islets and vascularization were greater in liver sections of mice receiving islets and FGF7-GAL-PLGA particles vs mice receiving islets alone, 72 h posttransplant. More mice (six of eight) that received islets and FGF7-GAL-PLGA particles normalized blood glucose concentrations by 30-days posttransplant, versus zero of eight mice receiving islets alone with no evidence of increased proliferation of cells within the liver at this stage and normal liver function tests. This work shows that liver-targeted FGF7-GAL-PLGA particles achieve selective FGF7 delivery to the liver-promoting islet engraftment to help normalize blood glucose levels with a good safety profile.
Collapse
Affiliation(s)
- Salamah M. Alwahsh
- Centre for Regenerative MedicineUniversity of EdinburghEdinburghUK,Joint MD ProgramCollege of Medicine and Health SciencesPalestine Polytechnic UniversityHebronPalestine
| | - Omar Qutachi
- School of PharmacyUniversity of NottinghamUniversity ParkNottinghamUK
| | | | - Andrew Bond
- BHF Centre for Cardiovascular ScienceUniversity of EdinburghQueen’s Medical Research InstituteEdinburghUK
| | - June Noble
- BHF Centre for Cardiovascular ScienceUniversity of EdinburghQueen’s Medical Research InstituteEdinburghUK
| | - Paul Burgoyne
- BHF Centre for Cardiovascular ScienceUniversity of EdinburghQueen’s Medical Research InstituteEdinburghUK
| | - Nik Morton
- BHF Centre for Cardiovascular ScienceUniversity of EdinburghQueen’s Medical Research InstituteEdinburghUK
| | - Rod Carter
- BHF Centre for Cardiovascular ScienceUniversity of EdinburghQueen’s Medical Research InstituteEdinburghUK
| | - Janet Mann
- Centre for Regenerative MedicineUniversity of EdinburghEdinburghUK
| | | | | | - Stuart J. Forbes
- Centre for Regenerative MedicineUniversity of EdinburghEdinburghUK
| | | | - Shareen Forbes
- BHF Centre for Cardiovascular ScienceUniversity of EdinburghQueen’s Medical Research InstituteEdinburghUK
| |
Collapse
|
9
|
Novel Coagulation Factor VIII Gene Therapy in a Mouse Model of Hemophilia A by Lipid-Coated Fe 3O 4 Nanoparticles. Biomedicines 2021; 9:biomedicines9091116. [PMID: 34572302 PMCID: PMC8464966 DOI: 10.3390/biomedicines9091116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/22/2021] [Accepted: 08/27/2021] [Indexed: 12/12/2022] Open
Abstract
Hemophilia A is a bleeding disease caused by loss of coagulation factor VIII (FVIII) function. Although prophylactic FVIII infusion prevents abnormal bleeding, disability and joint damage in hemophilia patients are common. The cost of treatment is among the highest for a single disease, and the adverse effects of repeated infusion are still an issue that has not been addressed. In this study, we established a nonviral gene therapy strategy to treat FVIII knockout (FVIII KO) mice. A novel gene therapy approach was developed using dipalmitoylphosphatidylcholine formulated with iron oxide (DPPC-Fe3O4) to carry the B-domain-deleted (BDD)-FVIII plasmid, which was delivered into the FVIII KO mice via tail vein injection. Here, a liver-specific albumin promoter-driven BDD-FVIII plasmid was constructed, and the binding ability of circular DNA was confirmed to be more stable than that of linear DNA when combined with DPPC-Fe3O4 nanoparticles. The FVIII KO mice that received the DPPC-Fe3O4 plasmid complex were assessed by staining the ferric ion of DPPC-Fe3O4 nanoparticles with Prussian blue in liver tissue. The bleeding of the FVIII KO mice was improved in a few weeks, as shown by assessing the activated partial thromboplastin time (aPTT). Furthermore, no liver toxicity, thromboses, deaths, or persistent changes after nonviral gene therapy were found, as shown by serum liver indices and histopathology. The results suggest that this novel gene therapy can successfully improve hemostasis disorder in FVIII KO mice and might be a promising approach to treating hemophilia A patients in clinical settings.
Collapse
|
10
|
Wang G, Zhang S, Lu H, Mu Y. Therapeutic Angiogenesis for Ovarian Transplantation through Ultrasound-Targeted Microbubble Destruction. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:1868-1880. [PMID: 33832825 DOI: 10.1016/j.ultrasmedbio.2021.02.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 02/25/2021] [Accepted: 02/28/2021] [Indexed: 06/12/2023]
Abstract
Timely angiogenesis and effective microcirculation perfusion are essential for the survival and functional recovery of transplanted ovaries. Ultrasound-targeted microbubble destruction (UTMD) can lead to angiogenesis and increase flow perfusion by causing transient inflammation. The purpose of this study was to evaluate the effects of UTMD on transplanted ovarian revascularization and survival. In vitro, for the criteria of cell viability and tube formation capability, the optimal exposure parameters were determined to be a microbubble concentration of 1 × 108/mL, mechanical index of 1 and exposure time of 30 s. After ovarian transplantation, 40 female Sprague Dawley rats were divided into four groups: transplantation alone, ultrasound alone, microbubbles alone and ultrasound and microbubbles (UTMD). At 7 d after transplantation, ovarian perfusion was assessed using qualitative and quantitative methods. The effect of angiogenesis was assessed by contrast-enhanced ultrasound, laser Doppler perfusion imaging and histologic analysis. The results, in which ovarian perfusion was highest in the UTMD group, suggest that UTMD can effectively improve ovarian perfusion. Compared with the other three groups, the number of follicles, microvascular density and rate of Ki-67-positive cells increased significantly in the UTMD group, while apoptosis decreased significantly (p < 0.05). The study indicates that UTMD promoted ovarian re-vascularization after ovarian transplantation and maintained follicular reserve.
Collapse
Affiliation(s)
- Guodong Wang
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Shan Zhang
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hanbing Lu
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yuming Mu
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
11
|
Jangjou A, Meisami AH, Jamali K, Niakan MH, Abbasi M, Shafiee M, Salehi M, Hosseinzadeh A, Amani AM, Vaez A. The promising shadow of microbubble over medical sciences: from fighting wide scope of prevalence disease to cancer eradication. J Biomed Sci 2021; 28:49. [PMID: 34154581 PMCID: PMC8215828 DOI: 10.1186/s12929-021-00744-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/10/2021] [Indexed: 12/29/2022] Open
Abstract
Microbubbles are typically 0.5-10 μm in size. Their size tends to make it easier for medication delivery mechanisms to navigate the body by allowing them to be swallowed more easily. The gas included in the microbubble is surrounded by a membrane that may consist of biocompatible biopolymers, polymers, surfactants, proteins, lipids, or a combination thereof. One of the most effective implementation techniques for tiny bubbles is to apply them as a drug carrier that has the potential to activate ultrasound (US); this allows the drug to be released by US. Microbubbles are often designed to preserve and secure medicines or substances before they have reached a certain area of concern and, finally, US is used to disintegrate microbubbles, triggering site-specific leakage/release of biologically active drugs. They have excellent therapeutic potential in a wide range of common diseases. In this article, we discussed microbubbles and their advantageous medicinal uses in the treatment of certain prevalent disorders, including Parkinson's disease, Alzheimer's disease, cardiovascular disease, diabetic condition, renal defects, and finally, their use in the treatment of various forms of cancer as well as their incorporation with nanoparticles. Using microbubble technology as a novel carrier, the ability to prevent and eradicate prevalent diseases has strengthened the promise of effective care to improve patient well-being and life expectancy.
Collapse
Affiliation(s)
- Ali Jangjou
- Department of Emergency Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Hossein Meisami
- Department of Emergency Medicine, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kazem Jamali
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hadi Niakan
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Abbasi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mostafa Shafiee
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Majid Salehi
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Ahmad Hosseinzadeh
- Thoracic and Vascular Surgery Research Center, Nemazee Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Amani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Vaez
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
Chowdhury SM, Abou-Elkacem L, Lee T, Dahl J, Lutz AM. Ultrasound and microbubble mediated therapeutic delivery: Underlying mechanisms and future outlook. J Control Release 2020; 326:75-90. [PMID: 32554041 DOI: 10.1016/j.jconrel.2020.06.008] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/06/2020] [Accepted: 06/09/2020] [Indexed: 12/20/2022]
Abstract
Beyond the emerging field of oncological ultrasound molecular imaging, the recent significant advancements in ultrasound and contrast agent technology have paved the way for therapeutic ultrasound mediated microbubble oscillation and has shown that this approach is capable of increasing the permeability of microvessel walls while also initiating enhanced extravasation and drug delivery into target tissues. In addition, a large number of preclinical studies have demonstrated that ultrasound alone or combined with microbubbles can efficiently increase cell membrane permeability resulting in enhanced tissue distribution and intracellular drug delivery of molecules, nanoparticles, and other therapeutic agents. The mechanism behind the enhanced permeability is the temporary creation of pores in cell membranes through a phenomenon called sonoporation by high-intensity ultrasound and microbubbles or cavitation agents. At low ultrasound intensities (0.3-3 W/cm2), sonoporation may be caused by microbubbles oscillating in a stable motion, also known as stable cavitation. In contrast, at higher ultrasound intensities (greater than 3 W/cm2), sonoporation usually occurs through inertial cavitation that accompanies explosive growth and collapse of the microbubbles. Sonoporation has been shown to be a highly effective method to improve drug uptake through microbubble potentiated enhancement of microvascular permeability. In this review, the therapeutic strategy of using ultrasound for improved drug delivery are summarized with the special focus on cancer therapy. Additionally, we discuss the progress, challenges, and future of ultrasound-mediated drug delivery towards clinical translation.
Collapse
Affiliation(s)
- Sayan Mullick Chowdhury
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA
| | - Lotfi Abou-Elkacem
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA
| | - Taehwa Lee
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeremy Dahl
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA
| | - Amelie M Lutz
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
13
|
Rademakers T, Horvath JM, van Blitterswijk CA, LaPointe VL. Oxygen and nutrient delivery in tissue engineering: Approaches to graft vascularization. J Tissue Eng Regen Med 2019; 13:1815-1829. [PMID: 31310055 PMCID: PMC6852121 DOI: 10.1002/term.2932] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/13/2019] [Accepted: 07/01/2019] [Indexed: 12/29/2022]
Abstract
The field of tissue engineering is making great strides in developing replacement tissue grafts for clinical use, marked by the rapid development of novel biomaterials, their improved integration with cells, better-directed growth and differentiation of cells, and improved three-dimensional tissue mass culturing. One major obstacle that remains, however, is the lack of graft vascularization, which in turn renders many grafts to fail upon clinical application. With that, graft vascularization has turned into one of the holy grails of tissue engineering, and for the majority of tissues, it will be imperative to achieve adequate vascularization if tissue graft implantation is to succeed. Many different approaches have been developed to induce or augment graft vascularization, both in vitro and in vivo. In this review, we highlight the importance of vascularization in tissue engineering and outline various approaches inspired by both biology and engineering to achieve and augment graft vascularization.
Collapse
Affiliation(s)
- Timo Rademakers
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Judith M. Horvath
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Clemens A. van Blitterswijk
- Complex Tissue Regeneration, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Vanessa L.S. LaPointe
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| |
Collapse
|
14
|
Chen Y, Nguyen DT, Kokil GR, Wong YX, Dang TT. Microencapsulated islet-like microtissues with toroid geometry for enhanced cellular viability. Acta Biomater 2019; 97:260-271. [PMID: 31404714 DOI: 10.1016/j.actbio.2019.08.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 08/08/2019] [Accepted: 08/08/2019] [Indexed: 12/27/2022]
Abstract
Transplantation of immuno-isolated islets is a promising strategy to restore insulin-secreting function in patients with Type 1 diabetes. However, the clinical translation of this treatment approach remains elusive due to the loss of islet viability resulting from hypoxia at the avascular transplantation site. To address this challenge, we designed non-spherical islet-like microtissues and investigated the effect of their geometries on cellular viability. Insulin-secreting microtissues with different shapes were fabricated by assembly of monodispersed rat insulinoma beta cells on micromolded nonadhesive hydrogels. Our study quantitatively demonstrated that toroid microtissues exhibited enhanced cellular viability and metabolic activity compared to rod and spheroid microtissues with the same volume. At a similar level of cellular viability, toroid geometry facilitated efficient packing of more cells into each microtissue than rod and spheroid geometries. In addition, toroid microtissues maintained the characteristic glucose-responsive insulin secretion of rat insulinoma beta cells. Furthermore, toroid microtissues preserved their geometry and structural integrity following their microencapsulation in immuno-isolatory alginate hydrogel. Our study suggests that adopting toroid geometry in designing therapeutic microtissues potentially reduces mass loss of cellular grafts and thereby may improve the performance of transplanted islets towards a clinically viable cure for Type 1 diabetes. STATEMENT OF SIGNIFICANCE: Transplantation of therapeutic cells is a promising strategy for the treatment of a wide range of hormone or protein-deficiency diseases. However, the clinical application of this approach is hindered by the loss of cell viability and function at the avascular transplantation site. To address this challenge, we fabricated hydrogel-encapsulated islet-like microtissues with non-spheroidal geometry and optimal surface-to-volume ratio. This study demonstrated that the viability of therapeutic cells can be significantly increased solely by redesigning the microtissue configuration without requiring any additional biochemical or operational accessories. This study suggests that the adoption of toroid geometry provides a possible avenue to improve the long-term survival of transplanted therapeutic cells and expedite the translation of cell-based therapy towards clinical application.
Collapse
Affiliation(s)
- Yang Chen
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore
| | - Dang T Nguyen
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore
| | - Ganesh R Kokil
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore
| | - Yun Xuan Wong
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore
| | - Tram T Dang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore.
| |
Collapse
|
15
|
Oakie A, Wang R. β-Cell Receptor Tyrosine Kinases in Controlling Insulin Secretion and Exocytotic Machinery: c-Kit and Insulin Receptor. Endocrinology 2018; 159:3813-3821. [PMID: 30239687 PMCID: PMC6202852 DOI: 10.1210/en.2018-00716] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 09/13/2018] [Indexed: 12/25/2022]
Abstract
Insulin secretion from pancreatic β-cells is initiated through channel-mediated depolarization, cytoskeletal remodeling, and vesicle tethering at the cell membrane, all of which can be regulated through cell surface receptors. Receptor tyrosine kinases (RTKs) promote β-cell development and postnatal signaling to improve β-cell mass and function, yet their activation has also been shown to initiate exocytotic events in β-cells. This review examines the role of RTK signaling in insulin secretion, with a focus on RTKs c-Kit and insulin receptor (IR). Pathways that control insulin release and the potential interplay between c-Kit and IR signaling are discussed, along with clinical implications of RTK therapy on insulin secretion.
Collapse
Affiliation(s)
- Amanda Oakie
- Children’s Health Research Institute, Victoria Research Laboratories, London, Ontario, Canada
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada
| | - Rennian Wang
- Children’s Health Research Institute, Victoria Research Laboratories, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
- Department of Medicine, University of Western Ontario, London, Ontario, Canada
- Correspondence: Rennian Wang, MD, PhD, Victoria Research Laboratories, Room A5-140, 800 Commissioners Road East, London, Ontario N6C 2V5, Canada. E-mail:
| |
Collapse
|
16
|
Abstract
PURPOSES OF REVIEW Scattered throughout the pancreas, the endocrine islets rely on neurovascular support for signal relay to regulate hormone secretion and for maintaining tissue homeostasis. The islet accessory cells (or components) of neurovascular tissues include the endothelial cells, pericytes, smooth muscle cells, neurons (nerve fibers), and glia. Research results derived from experimental diabetes and islet transplantation indicate that the accessory cells are reactive in islet injury and can affect islet function and homeostasis in situ or in an ectopic environment. RECENT FINDINGS Recent advances in cell labeling and tissue imaging have enabled investigation of islet accessory cells to gain insights into their network structures, functions, and remodeling in disease. It has become clear that in diabetes, the islet neurovascular tissues are not just bystanders damaged in neuropathy and vascular complications; rather, they participate in islet remodeling in response to changes in the microenvironment. Because of the fundamental differences between humans and animal models in neuroinsular cytoarchitecture and cell proliferation, examination of islet accessory cells in clinical specimens and donor pancreases warrants further attention.
Collapse
Affiliation(s)
- Shiue-Cheng Tang
- Department of Medical Science and Institute of Biotechnology, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| | - Claire F Jessup
- College of Medicine and Public Health, Flinders University and Discipline of Medicine, University of Adelaide, Adelaide, SA, 5001, Australia.
| | - Martha Campbell-Thompson
- Department of Pathology, Immunology, and Laboratory Medicine, 1395 Center Drive, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
17
|
ANGPTL8 reverses established adriamycin cardiomyopathy by stimulating adult cardiac progenitor cells. Oncotarget 2018; 7:80391-80403. [PMID: 27823982 PMCID: PMC5348328 DOI: 10.18632/oncotarget.13061] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 10/07/2016] [Indexed: 12/18/2022] Open
Abstract
Established adriamycin cardiomyopathy is a lethal disease. When congestive heart failure develops, mortality is approximately 50% in a year. It has been known that ANGPTLs has various functions in lipid metabolism, inflammation, cancer cell invasion, hematopoietic stem activity and diabetes. We hypothesized that ANGPTL8 is capable of maintaining heart function by stimulating adult cardiac progenitor cells to initiate myocardial regeneration. We employed UTMD to deliver piggybac transposon plasmids with the human ANGPTL8 gene to the liver of rats with adriamycin cardiomyopathy. After ANGPTL8 gene liver delivery, overexpression of transgenic human ANGPTL8 was found in rat liver cells and blood. UTMD- ANGPTL8 gene therapy restored LV mass, fractional shortening index, and LV posterior wall diameter to nearly normal. Our results also showed that ANGPTL8 reversed established ADM cardiomyopathy. This was associated with activation of ISL-1 positive cardiac progenitor cells in the epicardium. A time-course experiment shown that ISL-1 cardiac progenitor cells proliferated and formed a niche in the epicardial layer and then migrated into sub-epicardium. The observed myocardial regeneration accompanying reversal of adriamycin cardiomyopathy was associated with upregulation of PirB expression on the cell membrane of cardiac muscle cells or progenitor cells stimulated by ANGPTL8.
Collapse
|
18
|
A preclinical evaluation of alternative site for islet allotransplantation. PLoS One 2017; 12:e0174505. [PMID: 28358858 PMCID: PMC5373587 DOI: 10.1371/journal.pone.0174505] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 03/10/2017] [Indexed: 02/05/2023] Open
Abstract
The bone marrow cavity (BMC) has recently been identified as an alternative site to the liver for islet transplantation. This study aimed to compare the BMC with the liver as an islet allotransplantation site in diabetic monkeys. Diabetes was induced in Rhesus monkeys using streptozocin, and the monkeys were then divided into the following three groups: Group1 (islets transplanted in the liver with immunosuppressant), Group 2 (islets transplanted in the tibial BMC), and Group 3 (islets transplanted in the tibial BMC with immunosuppressant). The C-peptide and blood glucose levels were preoperatively measured. An intravenous glucose tolerance test (IVGTT) was conducted to assess graft function, and complete blood cell counts were performed to assess cell population changes. Cytokine expression was measured using an enzyme-linked immune sorbent assay (ELISA) and MILLIPLEX. Five monkeys in Group 3 exhibited a significantly increased insulin-independent time compared with the other groups (Group 1: 78.2 ± 19.0 days; Group 2: 58.8 ± 17.0 days; Group 3: 189.6 ± 26.2 days) and demonstrated increases in plasma C-peptide 4 months after transplantation. The infusion procedure was not associated with adverse effects. Functional islets in the BMC were observed 225 days after transplantation using the dithizone (DTZ) and insulin/glucagon stains. Our results showed that allogeneic islets transplanted in the BMC of diabetic Rhesus monkeys remained alive and functional for a longer time than those transplanted in the liver. This study was the first successful demonstration of allogeneic islet engraftment in the BMC of non-human primates (NHPs).
Collapse
|
19
|
Zhang L, Sun Z, Ren P, You M, Zhang J, Fang L, Wang J, Chen Y, Yan F, Zheng H, Xie M. Localized Delivery of shRNA against PHD2 Protects the Heart from Acute Myocardial Infarction through Ultrasound-Targeted Cationic Microbubble Destruction. Theranostics 2017; 7:51-66. [PMID: 28042316 PMCID: PMC5196885 DOI: 10.7150/thno.16074] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 08/30/2016] [Indexed: 01/22/2023] Open
Abstract
Hypoxia-inducible factor 1α (HIF-1α) plays a critical protective role in ischemic heart disease. Under normoxic conditions, HIF-1α was degraded by oxygen-dependent prolyl hydroxylase-2 (PHD2). Gene therapy has become a promising strategy to inhibit the degradation of HIF-1α and to improve cardiac function after ischemic injury. However, conventional gene delivery systems are difficult to achieve a targeted and localized gene delivery into the ischemic myocardia. Here, we report the localized myocardial delivery of shRNA against PHD2 through ultrasound-targeted microbubble destruction (UTMD) for protection the heart from acute myocardial infarction. In this study, a novel cationic microbubble was fabricated by using of the thin-film hydration and sonication method. The resulting microbubbles had a 28.2 ± 2.21 mV surface zeta potential and could greatly improve DNA binding performance, achieving 17.81 ± 1.46 μg of DNA loading capacity per 5 × 108 microbubbles. Combined with these cationic microbubbles, UTMD-mediated gene delivery was evaluated and the gene transfection efficiency was optimized in the H9C2 cardiac cells. Knockdown of PHD2 gene was successfully realized by UTMD-mediated shPHD2 transfection, resulting in HIF-1α-dependent protective effects on H9C2 cells through increasing the expression of HIF-1α, VEGF and bFGF. We further employed UTMD-mediated shPHD2 transfection into the localized ischemic myocardia in a rat ischemia model, demonstrating significantly reduced infarct size and greatly improved the heart function. The silencing of PHD2 and the up-regulation of its downstream genes in the treated myocardia were confirmed. Histological analysis further revealed numbers of HIF-1α- and VEGF-, and CD31-positive cells/mm2 in the shPHD2-treated group were significantly greater than those in the sham or control vector groups (P < 0.05). In conclusion, our study provides a promising strategy to realize ultrasound-mediated localized myocardial shRNA delivery to protect the heart from acute myocardial infarction via cationic microbubbles.
Collapse
|
20
|
|
21
|
Keeney M, Chung MT, Zielins ER, Paik KJ, McArdle A, Morrison SD, Ransom RC, Barbhaiya N, Atashroo D, Jacobson G, Zare RN, Longaker MT, Wan DC, Yang F. Scaffold-mediated BMP-2 minicircle DNA delivery accelerated bone repair in a mouse critical-size calvarial defect model. J Biomed Mater Res A 2016; 104:2099-107. [PMID: 27059085 DOI: 10.1002/jbm.a.35735] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 03/29/2016] [Accepted: 04/01/2016] [Indexed: 12/31/2022]
Abstract
Scaffold-mediated gene delivery holds great promise for tissue regeneration. However, previous attempts to induce bone regeneration using scaffold-mediated non-viral gene delivery rarely resulted in satisfactory healing. We report a novel platform with sustained release of minicircle DNA (MC) from PLGA scaffolds to accelerate bone repair. MC was encapsulated inside PLGA scaffolds using supercritical CO2 , which showed prolonged release of MC. Skull-derived osteoblasts transfected with BMP-2 MC in vitro result in higher osteocalcin gene expression and mineralized bone formation. When implanted in a critical-size mouse calvarial defect, scaffolds containing luciferase MC lead to robust in situ protein production up to at least 60 days. Scaffold-mediated BMP-2 MC delivery leads to substantially accelerated bone repair as early as two weeks, which continues to progress over 12 weeks. This platform represents an efficient, long-term nonviral gene delivery system, and may be applicable for enhancing repair of a broad range of tissues types. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 2099-2107, 2016.
Collapse
Affiliation(s)
- Michael Keeney
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Clark Center E-150, 300 Pasteur Drive, Edwards R105, MC5341, Stanford, California, 94305.,Department of Bioengineering, Stanford University School of Medicine, Clark Center E-150, 300 Pasteur Drive, Edwards R105, MC5341, Stanford, California, 94305
| | - Michael T Chung
- Department of Surgery, Plastic and Reconstructive Surgery Division, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, 257 Campus Drive, Stanford University, Stanford, California, 94305-5148
| | - Elizabeth R Zielins
- Department of Surgery, Plastic and Reconstructive Surgery Division, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, 257 Campus Drive, Stanford University, Stanford, California, 94305-5148
| | - Kevin J Paik
- Department of Surgery, Plastic and Reconstructive Surgery Division, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, 257 Campus Drive, Stanford University, Stanford, California, 94305-5148
| | - Adrian McArdle
- Department of Surgery, Plastic and Reconstructive Surgery Division, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, 257 Campus Drive, Stanford University, Stanford, California, 94305-5148
| | - Shane D Morrison
- Department of Surgery, Plastic and Reconstructive Surgery Division, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, 257 Campus Drive, Stanford University, Stanford, California, 94305-5148
| | - Ryan C Ransom
- Department of Surgery, Plastic and Reconstructive Surgery Division, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, 257 Campus Drive, Stanford University, Stanford, California, 94305-5148
| | - Namrata Barbhaiya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Clark Center E-150, 300 Pasteur Drive, Edwards R105, MC5341, Stanford, California, 94305.,Department of Bioengineering, Stanford University School of Medicine, Clark Center E-150, 300 Pasteur Drive, Edwards R105, MC5341, Stanford, California, 94305
| | - David Atashroo
- Department of Surgery, Plastic and Reconstructive Surgery Division, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, 257 Campus Drive, Stanford University, Stanford, California, 94305-5148
| | - Gunilla Jacobson
- Department of Chemistry, Stanford University, 333 Campus Drive Mudd Building, Room 121 Stanford, Stanford, California, 94305-4401
| | - Richard N Zare
- Department of Chemistry, Stanford University, 333 Campus Drive Mudd Building, Room 121 Stanford, Stanford, California, 94305-4401
| | - Michael T Longaker
- Department of Surgery, Plastic and Reconstructive Surgery Division, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, 257 Campus Drive, Stanford University, Stanford, California, 94305-5148
| | - Derrick C Wan
- Department of Surgery, Plastic and Reconstructive Surgery Division, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, 257 Campus Drive, Stanford University, Stanford, California, 94305-5148
| | - Fan Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Clark Center E-150, 300 Pasteur Drive, Edwards R105, MC5341, Stanford, California, 94305.,Department of Bioengineering, Stanford University School of Medicine, Clark Center E-150, 300 Pasteur Drive, Edwards R105, MC5341, Stanford, California, 94305
| |
Collapse
|
22
|
Mozafari M, Shimoda M, Urbanska AM, Laurent S. Ultrasound-targeted microbubble destruction: toward a new strategy for diabetes treatment. Drug Discov Today 2016; 21:540-3. [PMID: 26646254 DOI: 10.1016/j.drudis.2015.11.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 11/02/2015] [Accepted: 11/23/2015] [Indexed: 02/06/2023]
|
23
|
Xiao X, Fischbach S, Song Z, Gaffar I, Zimmerman R, Wiersch J, Prasadan K, Shiota C, Guo P, Ramachandran S, Witkowski P, Gittes GK. Transient Suppression of TGFβ Receptor Signaling Facilitates Human Islet Transplantation. Endocrinology 2016; 157:1348-1356. [PMID: 26872091 PMCID: PMC4816736 DOI: 10.1210/en.2015-1986] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/09/2016] [Indexed: 11/19/2022]
Abstract
Although islet transplantation is an effective treatment for severe diabetes, its broad application is greatly limited due to a shortage of donor islets. Suppression of TGFβ receptor signaling in β-cells has been shown to increase β-cell proliferation in mice, but has not been rigorously examined in humans. Here, treatment of human islets with a TGFβ receptor I inhibitor, SB-431542 (SB), significantly improved C-peptide secretion by β-cells, and significantly increased β-cell number by increasing β-cell proliferation. In addition, SB increased cell-cycle activators and decreased cell-cycle suppressors in human β-cells. Transplantation of SB-treated human islets into diabetic immune-deficient mice resulted in significant improvement in blood glucose control, significantly higher serum and graft insulin content, and significantly greater increases in β-cell proliferation in the graft, compared with controls. Thus, our data suggest that transient suppression of TGFβ receptor signaling may improve the outcome of human islet transplantation, seemingly through increasing β-cell number and function.
Collapse
Affiliation(s)
| | | | | | | | - Ray Zimmerman
- Division of Pediatric Surgery (X.X., S.F., Z.S., I.G., R.Z., J.W., K.P., C.S., P.G., G.K.G.), Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central S University, Changsha 410013, China; and Department of Surgery (S.R., P.W.), University of Chicago, Chicago, Illinois 60637
| | - John Wiersch
- Division of Pediatric Surgery (X.X., S.F., Z.S., I.G., R.Z., J.W., K.P., C.S., P.G., G.K.G.), Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central S University, Changsha 410013, China; and Department of Surgery (S.R., P.W.), University of Chicago, Chicago, Illinois 60637
| | - Krishna Prasadan
- Division of Pediatric Surgery (X.X., S.F., Z.S., I.G., R.Z., J.W., K.P., C.S., P.G., G.K.G.), Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central S University, Changsha 410013, China; and Department of Surgery (S.R., P.W.), University of Chicago, Chicago, Illinois 60637
| | - Chiyo Shiota
- Division of Pediatric Surgery (X.X., S.F., Z.S., I.G., R.Z., J.W., K.P., C.S., P.G., G.K.G.), Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central S University, Changsha 410013, China; and Department of Surgery (S.R., P.W.), University of Chicago, Chicago, Illinois 60637
| | - Ping Guo
- Division of Pediatric Surgery (X.X., S.F., Z.S., I.G., R.Z., J.W., K.P., C.S., P.G., G.K.G.), Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central S University, Changsha 410013, China; and Department of Surgery (S.R., P.W.), University of Chicago, Chicago, Illinois 60637
| | - Sabarinathan Ramachandran
- Division of Pediatric Surgery (X.X., S.F., Z.S., I.G., R.Z., J.W., K.P., C.S., P.G., G.K.G.), Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central S University, Changsha 410013, China; and Department of Surgery (S.R., P.W.), University of Chicago, Chicago, Illinois 60637
| | - Piotr Witkowski
- Division of Pediatric Surgery (X.X., S.F., Z.S., I.G., R.Z., J.W., K.P., C.S., P.G., G.K.G.), Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central S University, Changsha 410013, China; and Department of Surgery (S.R., P.W.), University of Chicago, Chicago, Illinois 60637
| | - George K. Gittes
- Division of Pediatric Surgery (X.X., S.F., Z.S., I.G., R.Z., J.W., K.P., C.S., P.G., G.K.G.), Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central S University, Changsha 410013, China; and Department of Surgery (S.R., P.W.), University of Chicago, Chicago, Illinois 60637
| |
Collapse
|
24
|
Chen J, Chen S, Huang P, Meng XL, Clayton S, Shen JS, Grayburn PA. In vivo targeted delivery of ANGPTL8 gene for beta cell regeneration in rats. Diabetologia 2015; 58:1036-44. [PMID: 25720603 DOI: 10.1007/s00125-015-3521-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/07/2015] [Indexed: 12/18/2022]
Abstract
AIMS/HYPOTHESIS ANGPTL8 is a circulatory hormone secreted from liver and adipose tissue that promotes pancreatic beta cell proliferation and interferes with triacylglycerol metabolism in mice. The clinical significance of its effects on inducing beta cell proliferation is limited because it causes severe hypertriacylglycerolaemia. METHODS We employed ultrasound-targeted microbubble destruction (UTMD) to deliver human ANGPTL8 gene plasmids to the pancreas, liver and skeletal muscle of normal adult rats. RESULTS Human ANGPTL8 was consistently detected in the circulation 1 month after UTMD. ANGPTL8 gene delivery promoted the proliferation of adult and aged beta cells, expanded the beta cell mass, improved glucose tolerance and increased the fasting blood insulin level after UTMD treatment without causing severe hypertriacylglycerolaemia. ANGPTL8 gene therapy significantly alleviated but did not totally reverse STZ-induced diabetes in a rat model. CONCLUSIONS/INTERPRETATION ANGPTL8 induced adult and aged beta cell regeneration in a rat model.
Collapse
Affiliation(s)
- Jiaxi Chen
- Medical School, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Phelps EA, Templeman KL, Thulé PM, García AJ. Engineered VEGF-releasing PEG-MAL hydrogel for pancreatic islet vascularization. Drug Deliv Transl Res 2015; 5:125-36. [PMID: 25787738 PMCID: PMC4366610 DOI: 10.1007/s13346-013-0142-2] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Biofunctionalized polyethylene glycol maleimide (PEG-MAL) hydrogels were engineered as a platform to deliver pancreatic islets to the small bowel mesentery and promote graft vascularization. VEGF, a potent stimulator of angiogenesis, was incorporated into the hydrogel to be released in an on-demand manner through enzymatic degradation. PEG-MAL hydrogel enabled extended in vivo release of VEGF. Isolated rat islets encapsulated in PEG-MAL hydrogels remained viable in culture and secreted insulin. Islets encapsulated in PEG-MAL matrix and transplanted to the small bowel mesentery of healthy rats grafted to the host tissue and revascularized by 4 weeks. Addition of VEGF release to the PEG-MAL matrix greatly augmented the vascularization response. These results establish PEG-MAL engineered matrices as a vascular-inductive cell delivery vehicle and warrant their further investigation as islet transplantation vehicles in diabetic animal models.
Collapse
Affiliation(s)
- Edward A. Phelps
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30332
| | - Kellie L. Templeman
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30332
| | - Peter M. Thulé
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30332
- Division of Endocrinology, Metabolism and Lipids, Emory University School of Medicine, Atlanta Veterans Affairs Medical Center, 1670 Clairmont Road NE, Decatur, GA 30033
| | - Andrés J. García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30332
| |
Collapse
|
26
|
Yang EY, Kronenfeld JP, Stabler CL. Engineering biomimetic materials for islet transplantation. Curr Diabetes Rev 2015; 11:163-9. [PMID: 25776871 PMCID: PMC4447569 DOI: 10.2174/1573399811666150317130440] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 12/30/2022]
Abstract
A closed-loop system that provides both the sensing of glucose and the appropriate dosage of insulin could dramatically improve treatment options for insulin-dependent diabetics. The intrahepatic implantation of allogeneic islets has the potential to provide this intimate control, by transplanting the very cells that have this inherent sensing and secretion capacity. Limiting islet transplantation, however, is the significant loss and dysfunction of islets following implantation, due to the poor engraftment environment and significant immunological attack. In this review, we outline approaches that seek to address these challenges via engineering biomimetic materials. These materials can serve to mimic natural processes that work toward improving engraftment, minimizing inflammation, and directing immunological responses. Biomimetic materials can serve to house cells, recapitulate native microenvironments, release therapeutic agents in a physiological manner, and/or present agents to direct cells towards desired responses. By integrating these approaches, superior platforms capable of improving long-term engraftment and acceptance of transplanted islets are on the horizon.
Collapse
Affiliation(s)
| | | | - Cherie L Stabler
- Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
27
|
Affiliation(s)
- Peter In't Veld
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium,
| | | |
Collapse
|
28
|
Abstract
The ultimate goal of diabetes therapy is the restoration of physiologic metabolic control. For type 1 diabetes, research efforts are focused on the prevention or early intervention to halt the autoimmune process and preserve β cell function. Replacement of pancreatic β cells via islet transplantation reestablishes physiologic β cell function in patients with diabetes. Emerging research shows that microRNAs (miRNAs), noncoding small RNA molecules produced by a newly discovered class of genes, negatively regulate gene expression. MiRNAs recognize and bind to partially complementary sequences of target messenger RNA (mRNA), regulating mRNA translation and affecting gene expression. Correlation between miRNA signatures and genome-wide RNA expression allows identification of multiple miRNA-mRNA pairs in biological processes. Because miRNAs target functionally related genes, they represent an exciting and indispensable approach for biomarkers and drug discovery. We are studying the role of miRNA in the context of islet immunobiology. Our research aims at understanding the mechanisms underlying pancreatic β cell loss and developing clinically relevant approaches for preservation and restoration of β cell function to treat insulin-dependent diabetes. Herein, we discuss some of our recent efforts related to the study of miRNA in islet inflammation and islet engraftment. Our working hypothesis is that modulation of the expression of specific microRNAs in the transplant microenvironment will be of assistance in enhancing islet engraftment and promoting long-term function.
Collapse
|
29
|
Unger E, Porter T, Lindner J, Grayburn P. Cardiovascular drug delivery with ultrasound and microbubbles. Adv Drug Deliv Rev 2014; 72:110-26. [PMID: 24524934 DOI: 10.1016/j.addr.2014.01.012] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Revised: 01/23/2014] [Accepted: 01/29/2014] [Indexed: 01/14/2023]
Abstract
Microbubbles lower the threshold for cavitation of ultrasound and have multiple potential therapeutic applications in the cardiovascular system. One of the first therapeutic applications to enter into clinical trials has been microbubble-enhanced sonothrombolysis. Trials were conducted in acute ischemic stroke and clinical trials are currently underway for sonothrombolysis in treatment of acute myocardial infarction. Microbubbles can be targeted to epitopes expressed on endothelial cells and thrombi by incorporating targeting ligands onto the surface of the microbubbles. Targeted microbubbles have applications as molecular imaging contrast agents and also for drug and gene delivery. A number of groups have shown that ultrasound with microbubbles can be used for gene delivery yielding robust gene expression in the target tissue. Work has progressed to primate studies showing delivery of therapeutic genes to generate islet cells in the pancreas to potentially cure diabetes. Microbubbles also hold potential as oxygen therapeutics and have shown promising results as a neuroprotectant in an ischemic stroke model. Regulatory considerations impact the successful clinical development of therapeutic applications of microbubbles with ultrasound. This paper briefly reviews the field and suggests avenues for further development.
Collapse
|
30
|
Shimoda M, Chen S, Noguchi H, Takita M, Sugimoto K, Itoh T, Chujo D, Iwahashi S, Naziruddin B, Levy MF, Matsumoto S, Grayburn PA. A new method for generating insulin-secreting cells from human pancreatic epithelial cells after islet isolation transformed by NeuroD1. Hum Gene Ther Methods 2014; 25:206-19. [PMID: 24845703 DOI: 10.1089/hgtb.2013.122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The generation of insulin-secreting cells from nonendocrine pancreatic epithelial cells (NEPEC) has been demonstrated for potential clinical use in the treatment of diabetes. However, previous methods either had limited efficacy or required viral vectors, which hinder clinical application. In this study, we aimed to establish an efficient method of insulin-secreting cell generation from NEPEC without viral vectors. We used nonislet fractions from both research-grade human pancreata from brain-dead donors and clinical pancreata after total pancreatectomy with autologous islet transplantation to treat chronic pancreatitis. It is of note that a few islets could be mingled in the nonislet fractions, but their influence could be limited. The NeuroD1 gene was induced into NEPEC using an effective triple lipofection method without viral vectors to generate insulin-secreting cells. The differentiation was promoted by adding a growth factor cocktail into the culture medium. Using the research-grade human pancreata, the effective method showed high efficacy in the differentiation of NEPEC into insulin-positive cells that secreted insulin in response to a glucose challenge and improved diabetes after being transplanted into diabetic athymic mice. Using the clinical pancreata, similar efficacy was obtained, even though those pancreata suffered chronic pancreatitis. In conclusion, our effective differentiation protocol with triple lipofection method enabled us to achieve very efficient insulin-secreting cell generation from human NEPEC without viral vectors. This method offers the potential for supplemental insulin-secreting cell transplantation for both allogeneic and autologous islet transplantation.
Collapse
|
31
|
Xiao X, Prasadan K, Guo P, El-Gohary Y, Fischbach S, Wiersch J, Gaffar I, Shiota C, Gittes GK. Pancreatic duct cells as a source of VEGF in mice. Diabetologia 2014; 57:991-1000. [PMID: 24535231 PMCID: PMC3986695 DOI: 10.1007/s00125-014-3179-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 01/16/2014] [Indexed: 12/25/2022]
Abstract
AIMS/HYPOTHESIS Vascular endothelial growth factor (VEGF) is essential for proper pancreatic development, islet vascularisation and insulin secretion. In the adult pancreas, VEGF is thought to be predominantly secreted by beta cells. Although human duct cells have previously been shown to secrete VEGF at angiogenic levels in culture, an analysis of the kinetics of VEGF synthesis and secretion, as well as elucidation of an in vivo role for this ductal VEGF in affecting islet function and physiology, has been lacking. METHODS We analysed purified duct cells independently prepared by flow cytometry, surgical isolation or laser-capture microdissection. We infected duct cells in vivo with Vegf (also known as Vegfa) short hairpin RNA (shRNA) in an intrapancreatic ductal infusion system and examined the effect of VEGF knockdown in duct cells in vitro and in vivo. RESULTS Pancreatic duct cells express high levels of Vegf mRNA. Compared with beta cells, duct cells had a much higher ratio of secreted to intracellular VEGF. As a bioassay, formation of tubular structures by human umbilical vein endothelial cells was essentially undetectable when cultured alone and was substantially increased when co-cultured with pancreatic duct cells but significantly reduced when co-cultured with duct cells pretreated with Vegf shRNA. Compared with islets transplanted alone, improved vascularisation and function was detected in the islets co-transplanted with duct cells but not in islets co-transplanted with duct cells pretreated with Vegf shRNA. CONCLUSIONS/INTERPRETATION Human islet preparations for transplantation typically contain some contaminating duct cells and our findings suggest that the presence of duct cells in the islet preparation may improve transplantation outcomes.
Collapse
Affiliation(s)
- Xiangwei Xiao
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Ave, Pittsburgh, PA, 15224, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Huang L, Belousova T, Pan JSC, Du J, Ju H, Lu L, Zhang P, Truong LD, Nuotio-Antar A, Sheikh-Hamad D. AKI after conditional and kidney-specific knockdown of stanniocalcin-1. J Am Soc Nephrol 2014; 25:2303-15. [PMID: 24700878 DOI: 10.1681/asn.2013070690] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Stanniocalcin-1 is an intracrine protein; it binds to the cell surface, is internalized to the mitochondria, and diminishes superoxide generation through induction of uncoupling proteins. In vitro, stanniocalcin-1 inhibits macrophages and preserves endothelial barrier function, and transgenic overexpression of stanniocalcin-1 in mice protects against ischemia-reperfusion kidney injury. We sought to determine the kidney phenotype after kidney endothelium-specific expression of stanniocalcin-1 small hairpin RNA (shRNA). We generated transgenic mice that express stanniocalcin-1 shRNA or scrambled shRNA upon removal of a floxed reporter (phosphoglycerate kinase-driven enhanced green fluorescent protein) and used ultrasound microbubbles to deliver tyrosine kinase receptor-2 promoter-driven Cre to the kidney to permit kidney endothelium-specific shRNA expression. Stanniocalcin-1 mRNA and protein were expressed throughout the kidney in wild-type mice. Delivery of tyrosine kinase receptor-2 promoter-driven Cre to stanniocalcin-1 shRNA transgenic kidneys diminished the expression of stanniocalcin-1 mRNA and protein throughout the kidneys. Stanniocalcin-1 mRNA and protein expression did not change in similarly treated scrambled shRNA transgenic kidneys, and we observed no Cre protein expression in cultured and tyrosine kinase receptor-2 promoter-driven Cre-transfected proximal tubule cells, suggesting that knockdown of stanniocalcin-1 in epithelial cells in vivo may result from stanniocalcin-1 shRNA transfer from endothelial cells to epithelial cells. Kidney-specific knockdown of stanniocalcin-1 led to severe proximal tubule injury characterized by vacuolization, decreased uncoupling of protein-2 expression, greater generation of superoxide, activation of the unfolded protein response, initiation of autophagy, cell apoptosis, and kidney failure. Our observations suggest that stanniocalcin-1 is critical for tubular epithelial survival under physiologic conditions.
Collapse
Affiliation(s)
| | | | | | - Jie Du
- Department of Medicine/Division of Nephrology
| | - Huiming Ju
- Department of Medicine/Division of Nephrology
| | - Lianghao Lu
- Department of Medicine/Division of Nephrology
| | - Pumin Zhang
- Department of Molecular Physiology and Biophysics, and
| | - Luan D Truong
- Kidney Pathology Laboratory, The Methodist Hospital/Weill Cornell University, Houston, Texas
| | - Alli Nuotio-Antar
- Department of Pediatrics/Nutrition, Baylor College of Medicine, Houston, Texas; and
| | | |
Collapse
|
33
|
Chhabra P, Brayman KL. Overcoming barriers in clinical islet transplantation: current limitations and future prospects. Curr Probl Surg 2014; 51:49-86. [PMID: 24411187 DOI: 10.1067/j.cpsurg.2013.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
34
|
Peiris H, Bonder CS, Coates PTH, Keating DJ, Jessup CF. The β-cell/EC axis: how do islet cells talk to each other? Diabetes 2014; 63:3-11. [PMID: 24357688 DOI: 10.2337/db13-0617] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Within the pancreatic islet, the β-cell represents the ultimate biosensor. Its central function is to accurately sense glucose levels in the blood and consequently release appropriate amounts of insulin. As the only cell type capable of insulin production, the β-cell must balance this crucial workload with self-preservation and, when required, regeneration. Evidence suggests that the β-cell has an important ally in intraislet endothelial cells (ECs). As well as providing a conduit for delivery of the primary input stimulus (glucose) and dissemination of its most important effector (insulin), intraislet blood vessels deliver oxygen to these dense clusters of metabolically active cells. Furthermore, it appears that ECs directly impact insulin gene expression and secretion and β-cell survival. This review discusses the molecules and pathways involved in the crosstalk between β-cells and intraislet ECs. The evidence supporting the intraislet EC as an important partner for β-cell function is examined to highlight the relevance of this axis in the context of type 1 and type 2 diabetes. Recent work that has established the potential of ECs or their progenitors to enhance the re-establishment of glycemic control following pancreatic islet transplantation in animal models is discussed.
Collapse
Affiliation(s)
- Heshan Peiris
- Department of Human Physiology, Centre for Neuroscience, Flinders University of South Australia, Adelaide, Australia
| | | | | | | | | |
Collapse
|
35
|
Brady AC, Martino MM, Pedraza E, Sukert S, Pileggi A, Ricordi C, Hubbell JA, Stabler CL. Proangiogenic hydrogels within macroporous scaffolds enhance islet engraftment in an extrahepatic site. Tissue Eng Part A 2013; 19:2544-52. [PMID: 23790218 DOI: 10.1089/ten.tea.2012.0686] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The transplantation of allogeneic islets in recent clinical trials has shown substantial promise as a therapy for type 1 diabetes; however, long-term insulin independence remains inadequate. This has been largely attributed to the current intravascular, hepatic transplant site, which exposes islets to mechanical and inflammatory stresses. A highly macroporous scaffold, housed within an alternative transplant site, can support an ideal environment for islet transplantation by providing three-dimensional distribution of islets, while permitting the infiltration of host vasculature. In the present study, we sought to evaluate the synergistic effect of a proangiogenic hydrogel loaded within the void space of a macroporous poly(dimethylsiloxane) (PDMS) scaffold on islet engraftment. The fibrin-based proangiogenic hydrogel tested presents platelet derived growth factor (PDGF-BB), via a fibronectin (FN) fragment containing growth factor and major integrin binding sites in close proximity. The combination of the proangiogenic hydrogel with PDMS scaffolds resulted in a significant decrease in the time to normoglycemia for syngeneic mouse islet transplants. This benefit was associated with an observed increase in competent vessel branching, as well as mature intraislet vessels. Overall, the addition of the proangiogenic factor PDGF-BB, delivered via the FN fragment-functionalized hydrogel, positively influenced the efficiency of engraftment. These characteristics, along with its ease of retrieval, make this combination of a biostable macroporous scaffold and a degradable proangiogenic hydrogel a supportive structure for insulin-producing cells implanted in extrahepatic sites.
Collapse
|
36
|
Coronel MM, Stabler CL. Engineering a local microenvironment for pancreatic islet replacement. Curr Opin Biotechnol 2013; 24:900-8. [PMID: 23769320 DOI: 10.1016/j.copbio.2013.05.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 05/16/2013] [Accepted: 05/17/2013] [Indexed: 01/10/2023]
Abstract
Intraportal islet transplantation has emerged as a promising treatment for type 1 diabetes mellitus (T1DM). Nevertheless, long-term efficacy has been limited to a marginal number of patients. Outcomes have been restricted, in part, by challenges associated with the transplant site, poor vascularization, and disruption of the native islet architecture during the isolation process. Engineering a biomaterial platform that recapitulates critical components of the pancreatic environment can serve to address these hurdles. This review highlights the challenges and opportunities in engineering 3D niches for islets, specifically: the importance of site selection; the application of scaffold functionalization to present bioactive motifs; and the development of technologies for enhancing implant nutritional profiles. The potential of these novel approaches to improve islet engraftment and duration of function is discussed.
Collapse
Affiliation(s)
- Maria M Coronel
- Department of Biomedical Engineering, College of Engineering, University of Miami, Coral Gables, FL 33146, United States; Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | | |
Collapse
|
37
|
Yang D, Tan KB, Gao YH, Liu H, Yang WX. Effects of diagnostic ultrasound-targeted microbubble destruction on permeability of normal liver in rats. ULTRASONICS 2012; 52:1065-1071. [PMID: 23021237 DOI: 10.1016/j.ultras.2012.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 08/22/2012] [Accepted: 09/01/2012] [Indexed: 06/01/2023]
Abstract
This work investigated the effect of diagnostic ultrasound-targeted microbubble destruction (UTMD) on the permeability of normal liver tissue and the safety of this technique. One hundred and four rats were divided into four groups: the control group, the microbubble-only (MB) group, the ultrasound-only (US) group, and the ultrasound-targeted microbubble destruction group (UTMD). The permeabilities of capillaries and cell membranes were determined using Evans blue and lanthanum nitrate as tracers, respectively. The amount of Evans blue was approximately fourfold higher in the UTMD group than in the control, MB-only, and US-only groups (all P<0.01). Evans blue extravasation, visualized as red fluorescence, was detectable by laser confocal scanning microscopy in the parenchyma only in the UTMD group. Lanthanum nitrate-tracing transmission electron microscopy examination indicated that intracellular lanthanum was detectable in the cytoplasm only in the UTMD group. Blood chemical analysis indicated that the effect of diagnostic ultrasound-targeted microbubble destruction on the rats' serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels was transient and recoverable and that this technique had no obvious effect on renal function. Cellular swelling was observed in liver cells in the UTMD group at 0.5 h, but this swelling was no longer apparent after 1 week. These results suggest that diagnostic ultrasound-targeted microbubble destruction can increase the capillary and cell membrane permeabilities in normal liver tissue without a significant increase in hepatic and renal toxicity.
Collapse
Affiliation(s)
- Dan Yang
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, No. 183 Xinqiao Street, Chongqing 400037, China.
| | | | | | | | | |
Collapse
|
38
|
Monsma DJ, Monks NR, Cherba DM, Dylewski D, Eugster E, Jahn H, Srikanth S, Scott SB, Richardson PJ, Everts RE, Ishkin A, Nikolsky Y, Resau JH, Sigler R, Nickoloff BJ, Webb CP. Genomic characterization of explant tumorgraft models derived from fresh patient tumor tissue. J Transl Med 2012; 10:125. [PMID: 22709571 PMCID: PMC3439334 DOI: 10.1186/1479-5876-10-125] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 06/18/2012] [Indexed: 12/11/2022] Open
Abstract
Background There is resurgence within drug and biomarker development communities for the use of primary tumorgraft models as improved predictors of patient tumor response to novel therapeutic strategies. Despite perceived advantages over cell line derived xenograft models, there is limited data comparing the genotype and phenotype of tumorgrafts to the donor patient tumor, limiting the determination of molecular relevance of the tumorgraft model. This report directly compares the genomic characteristics of patient tumors and the derived tumorgraft models, including gene expression, and oncogenic mutation status. Methods Fresh tumor tissues from 182 cancer patients were implanted subcutaneously into immune-compromised mice for the development of primary patient tumorgraft models. Histological assessment was performed on both patient tumors and the resulting tumorgraft models. Somatic mutations in key oncogenes and gene expression levels of resulting tumorgrafts were compared to the matched patient tumors using the OncoCarta (Sequenom, San Diego, CA) and human gene microarray (Affymetrix, Santa Clara, CA) platforms respectively. The genomic stability of the established tumorgrafts was assessed across serial in vivo generations in a representative subset of models. The genomes of patient tumors that formed tumorgrafts were compared to those that did not to identify the possible molecular basis to successful engraftment or rejection. Results Fresh tumor tissues from 182 cancer patients were implanted into immune-compromised mice with forty-nine tumorgraft models that have been successfully established, exhibiting strong histological and genomic fidelity to the originating patient tumors. Comparison of the transcriptomes and oncogenic mutations between the tumorgrafts and the matched patient tumors were found to be stable across four tumorgraft generations. Not only did the various tumors retain the differentiation pattern, but supporting stromal elements were preserved. Those genes down-regulated specifically in tumorgrafts were enriched in biological pathways involved in host immune response, consistent with the immune deficiency status of the host. Patient tumors that successfully formed tumorgrafts were enriched for cell signaling, cell cycle, and cytoskeleton pathways and exhibited evidence of reduced immunogenicity. Conclusions The preservation of the patient’s tumor genomic profile and tumor microenvironment supports the view that primary patient tumorgrafts provide a relevant model to support the translation of new therapeutic strategies and personalized medicine approaches in oncology.
Collapse
Affiliation(s)
- David J Monsma
- Laboratory for Translational Medicine, Van Andel Research Institute, Grand Rapids, MI, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Saito Y, Chan NK, Sakata N, Hathout E. Nerve growth factor is associated with islet graft failure following intraportal transplantation. Islets 2012; 4:24-31. [PMID: 22192949 PMCID: PMC3365801 DOI: 10.4161/isl.18467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Nerve growth factor (NGF) has recently been recognized as an angiogenic factor with an important regulatory role in pancreatic β-cell function. We previously showed that treatment of pancreatic islets with NGF improved their quality and viability. Revascularization and survival of islets transplanted under the kidney capsule were improved by NGF. However, the usefulness of NGF in intraportal islet transplantation was not previously tested. To resolve this problem, we transplanted syngeneic islets (360 islet equivalents per recipient) cultured with or without NGF into the portal vein of streptozotocin-induced diabetic BALB/c mice. Analysis revealed that 44.4% (4/9) of control and 12.5% (1/8) of NGF-treated mice attained normoglycemia (≤ 200 mg/dL) (p = 0.195). NGF-treated islets led to worse graft function (area under the curve of intraperitoneal glucose tolerance tests (IPGTT) on post-operative day (POD) 30, control; 35,800 ± 3,960 min*mg/dl, NGF-treated; 47,900 ± 3,220 min*mg/dl: *p = 0.0348). NGF treatment of islets was also associated with increased graft failure [the percentage of TdT-mediated dUTP-biotin nick-end labeling (TUNEL)-positive and necrotic transplanted islets on POD 5, control; 23.8% (5/21), NGF-treated; 52.9% (9/17): p = 0.0650] following intraportal islet transplantation. Nonviable (TUNEL-positive and necrotic) islets in both groups expressed vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1α (HIF-1α). On the other hand, viable (TUNEL-negative and not necrotic) islets in both groups did not express VEGF and HIF-1α. In the present study, pre-transplant NGF treatment was associated with impaired survival and angiogenesis of intraportal islet grafts. The effect of NGF on islet transplantation may significantly vary according to the transplant site.
Collapse
Affiliation(s)
- Yukihiko Saito
- Islet Transplant Laboratory; Department of Pediatrics; Loma Linda University School of Medicine; Loma Linda, CA USA
- Division of Advanced Surgical Science and Technology; Department of Surgery; Tohoku University; Sendai, Japan
| | - Nathaniel K. Chan
- Islet Transplant Laboratory; Department of Pediatrics; Loma Linda University School of Medicine; Loma Linda, CA USA
| | - Naoaki Sakata
- Division of Hepato-Biliary Pancreatic Surgery; Department of Surgery; Tohoku University; Sendai, Japan
| | - Eba Hathout
- Islet Transplant Laboratory; Department of Pediatrics; Loma Linda University School of Medicine; Loma Linda, CA USA
- * Correspondence to: Eba Hathout;
| |
Collapse
|
40
|
Chhabra P, Brayman KL. Current status of immunomodulatory and cellular therapies in preclinical and clinical islet transplantation. J Transplant 2011; 2011:637692. [PMID: 22046502 PMCID: PMC3199196 DOI: 10.1155/2011/637692] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Accepted: 07/11/2011] [Indexed: 02/08/2023] Open
Abstract
Clinical islet transplantation is a β-cell replacement strategy that represents a possible definitive intervention for patients with type 1 diabetes, offering substantial benefits in terms of lowering daily insulin requirements and reducing incidences of debilitating hypoglycemic episodes and unawareness. Despite impressive advances in this field, a limiting supply of islets, inadequate means for preventing islet rejection, and the deleterious diabetogenic and nephrotoxic side effects associated with chronic immunosuppressive therapy preclude its wide-spread applicability. Islet transplantation however allows a window of opportunity for attempting various therapeutic manipulations of islets prior to transplantation aimed at achieving superior transplant outcomes. In this paper, we will focus on the current status of various immunosuppressive and cellular therapies that promote graft function and survival in preclinical and clinical islet transplantation with special emphasis on the tolerance-inducing capacity of regulatory T cells as well as the β-cells regenerative capacity of stem cells.
Collapse
Affiliation(s)
- Preeti Chhabra
- Department of Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Kenneth L. Brayman
- Department of Surgery, University of Virginia, Charlottesville, VA 22908, USA
- Division of Transplantation, Department of Surgery, University of Virginia, Charlottesville, VA 22908, USA
- The Center for Cellular Transplantation and Therapeutics, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
41
|
Jahansouz C, Jahansouz C, Kumer SC, Brayman KL. Evolution of β-Cell Replacement Therapy in Diabetes Mellitus: Islet Cell Transplantation. J Transplant 2011; 2011:247959. [PMID: 22013505 PMCID: PMC3195999 DOI: 10.1155/2011/247959] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Accepted: 08/08/2011] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus remains one of the leading causes of morbidity and mortality worldwide. According to the Centers for Disease Control and Prevention, approximately 23.6 million people in the United States are affected. Of these individuals, 5 to 10% have been diagnosed with Type 1 diabetes mellitus (T1DM), an autoimmune disease. Although it often appears in childhood, T1DM may manifest at any age, leading to significant morbidity and decreased quality of life. Since the 1960s, the surgical treatment for diabetes mellitus has evolved to become a viable alternative to insulin administration, beginning with pancreatic transplantation. While islet cell transplantation has emerged as another potential alternative, its role in the treatment of T1DM remains to be solidified as research continues to establish it as a truly viable alternative for achieving insulin independence. In this paper, the historical evolution, procurement, current status, benefits, risks, and ongoing research of islet cell transplantation are explored.
Collapse
Affiliation(s)
- Cyrus Jahansouz
- School of Medicine, University of Virginia, Charlottesville, VA 22102, USA
| | | | | | | |
Collapse
|
42
|
Walton CB, Anderson CD, Boulay R, Shohet RV. Introduction to the ultrasound targeted microbubble destruction technique. J Vis Exp 2011:2963. [PMID: 21694693 DOI: 10.3791/2963] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In UTMD, bioactive molecules, such as negatively charged plasmid DNA vectors encoding a gene of interest, are added to the cationic shells of lipid microbubble contrast agents. In mice these vector-carrying microbubbles can be administered intravenously or directly to the left ventricle of the heart. In larger animals they can also be infused through an intracoronary catheter. The subsequent delivery from the circulation to a target organ occurs by acoustic cavitation at a resonant frequency of the microbubbles. It seems likely that the mechanical energy generated by the microbubble destruction results in transient pore formation in or between the endothelial cells of the microvasculature of the targeted region. As a result of this sonoporation effect, the transfection efficiency into and across the endothelial cells is enhanced, and transgene-encoding vectors are deposited into the surrounding tissue. Plasmid DNA remaining in the circulation is rapidly degraded by nucleases in the blood, which further reduces the likelihood of delivery to non-sonicated tissues and leads to highly specific target-organ transfection.
Collapse
Affiliation(s)
- Chad B Walton
- Department of Medicine, JABSOM, University of Hawaii, USA
| | | | | | | |
Collapse
|
43
|
Lau J, Zang G, Carlsson PO. Pancreatic islet transplantation to the liver: how can vascularization problems be resolved? ACTA ACUST UNITED AC 2011. [DOI: 10.2217/dmt.11.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
44
|
Abstract
Pancreatic islet transplantation into the liver provides a possibility to treat selected patients with brittle type 1 diabetes mellitus. However, massive early β-cell death increases the number of islets needed to restore glucose homeostasis. Moreover, late dysfunction and death contribute to the poor long-term results of islet transplantation on insulin independence. Studies in recent years have identified early and late challenges for transplanted pancreatic islets, including an instant blood-mediated inflammatory reaction when exposing human islets to the blood microenvironment in the portal vein and the low oxygenated milieu of islets transplanted into the liver. Poor revascularization of remaining intact islets combined with severe changes in the gene expression of islets transplanted into the liver contributes to late dysfunction. Strategies to overcome these hurdles have been developed, and some of these interventions are now even tested in clinical trials providing a hope to improve results in clinical islet transplantation. In parallel, experimental and clinical studies have, based on the identified problems with the liver site, evaluated the possibility of change of implantation organ in order to improve the results. Site-specific differences clearly exist in the engraftment of transplanted islets, and a more thorough characterization of alternative locations is needed. New strategies with modifications of islet microenvironment with cells and growth factors adhered to the islet surface or in a surrounding matrix could be designed to intervene with site-specific hurdles and provide possibilities to improve future results of islet transplantation.
Collapse
Affiliation(s)
- Per-Ola Carlsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|