1
|
Mbaye EHA, Scott EA, Burke JA. From Edmonton to Lantidra and beyond: immunoengineering islet transplantation to cure type 1 diabetes. FRONTIERS IN TRANSPLANTATION 2025; 4:1514956. [PMID: 40182604 PMCID: PMC11965681 DOI: 10.3389/frtra.2025.1514956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025]
Abstract
Type 1 diabetes (T1D) is characterized by the autoimmune destruction of insulin-producing β cells within pancreatic islets, the specialized endocrine cell clusters of the pancreas. Islet transplantation has emerged as a β cell replacement therapy, involving the infusion of cadaveric islets into a patient's liver through the portal vein. This procedure offers individuals with T1D the potential to restore glucose control, reducing or even eliminating the need for exogenous insulin therapy. However, it does not address the underlying autoimmune condition responsible for T1D. The need for systemic immunosuppression remains the primary barrier to making islet transplantation a more widespread therapy for patients with T1D. Here, we review recent progress in addressing the key limitations of islet transplantation as a viable treatment for T1D. Concerns over systemic immunosuppression arise from its potential to cause severe side effects, including opportunistic infections, malignancies, and toxicity to transplanted islets. Recognizing the risks, the Edmonton protocol (2000) marked a shift away from glucocorticoids to prevent β cell damage specifically. This transition led to the development of combination immunosuppressive therapies and the emergence of less toxic immunosuppressive and anti-inflammatory drugs. More recent advances in islet transplantation derive from islet encapsulation devices, biomaterial platforms releasing immunomodulatory compounds or surface-modified with immune regulating ligands, islet engineering and co-transplantation with accessory cells. While most of the highlighted studies in this review remain at the preclinical stage using mouse and non-human primate models, they hold significant potential for clinical translation if a transdisciplinary research approach is prioritized.
Collapse
Affiliation(s)
- El Hadji Arona Mbaye
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, United States
| | - Evan A. Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, United States
- Department of Biomedical Engineering, NanoSTAR Institute, University of Virginia School of Medicine, Charlottesville, VA, United States
| | | |
Collapse
|
2
|
O’Brien J, Niehaus P, Chang K, Remark J, Barrett J, Dasgupta A, Adenegan M, Salimian M, Kevas Y, Chandrasekaran K, Kristian T, Chellappan R, Rubin S, Kiemen A, Lu CPJ, Russell JW, Ho CY. Skin keratinocyte-derived SIRT1 and BDNF modulate mechanical allodynia in mouse models of diabetic neuropathy. Brain 2024; 147:3471-3486. [PMID: 38554393 PMCID: PMC11449144 DOI: 10.1093/brain/awae100] [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: 10/05/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 04/01/2024] Open
Abstract
Diabetic neuropathy is a debilitating disorder characterized by spontaneous and mechanical allodynia. The role of skin mechanoreceptors in the development of mechanical allodynia is unclear. We discovered that mice with diabetic neuropathy had decreased sirtuin 1 (SIRT1) deacetylase activity in foot skin, leading to reduced expression of brain-derived neurotrophic factor (BDNF) and subsequent loss of innervation in Meissner corpuscles, a mechanoreceptor expressing the BDNF receptor TrkB. When SIRT1 was depleted from skin, the mechanical allodynia worsened in diabetic neuropathy mice, likely due to retrograde degeneration of the Meissner-corpuscle innervating Aβ axons and aberrant formation of Meissner corpuscles which may have increased the mechanosensitivity. The same phenomenon was also noted in skin-keratinocyte specific BDNF knockout mice. Furthermore, overexpression of SIRT1 in skin induced Meissner corpuscle reinnervation and regeneration, resulting in significant improvement of diabetic mechanical allodynia. Overall, the findings suggested that skin-derived SIRT1 and BDNF function in the same pathway in skin sensory apparatus regeneration and highlighted the potential of developing topical SIRT1-activating compounds as a novel treatment for diabetic mechanical allodynia.
Collapse
Affiliation(s)
- Jennifer O’Brien
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Peter Niehaus
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Koping Chang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, National Taiwan University, Taipei, 100, Taiwan
| | - Juliana Remark
- Hansjörg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Joy Barrett
- Hansjörg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Abhishikta Dasgupta
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Morayo Adenegan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Mohammad Salimian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yanni Kevas
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Krish Chandrasekaran
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - Tibor Kristian
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21021, USA
| | - Rajeshwari Chellappan
- Department of Pathology, University of Alabama Birmingham, Birmingham, AL 35233, USA
| | - Samuel Rubin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Chemistry, College of William and Mary, Williamsburg, VA 23187, USA
| | - Ashley Kiemen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Catherine Pei-Ju Lu
- Hansjörg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - James W Russell
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - Cheng-Ying Ho
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
3
|
Pilz J, Gloddek N, Lindheimer F, Lindner MJ, Puhr-Westerheide D, Ümütlü M, Cyran C, Seidensticker M, Lindner R, Kraetzl M, Renner S, Merkus D, Teupser D, Bartenstein P, Ziegler SI, Wolf E, Kemter E. Functional maturation and longitudinal imaging of intraportal neonatal porcine islet grafts in genetically diabetic pigs. Am J Transplant 2024; 24:1395-1405. [PMID: 38432328 DOI: 10.1016/j.ajt.2024.02.026] [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: 12/19/2023] [Revised: 02/07/2024] [Accepted: 02/24/2024] [Indexed: 03/05/2024]
Abstract
Allogeneic intraportal islet transplantation (ITx) has become an established treatment for patients with poorly controlled type 1 diabetes. However, the loss of viable beta-cell mass after transplantation remains a major challenge. Therefore, noninvasive imaging methods for long-term monitoring of the transplant fate are required. In this study, [68Ga]Ga-DOTA-exendin-4 positron emission tomography/computed tomography (PET/CT) was used for repeated monitoring of allogeneic neonatal porcine islets (NPI) after intraportal transplantation into immunosuppressed genetically diabetic pigs. NPI transplantation (3320-15,000 islet equivalents per kg body weight) led to a reduced need for exogenous insulin therapy and finally normalization of blood glucose levels in 3 out of 4 animals after 5 to 10 weeks. Longitudinal PET/CT measurements revealed a significant increase in standard uptake values in graft-bearing livers. Histologic analysis confirmed the presence of well-engrafted, mature islet clusters in the transplanted livers. Our study presents a novel large animal model for allogeneic intraportal ITx. A relatively small dose of NPIs was sufficient to normalize blood glucose levels in a clinically relevant diabetic pig model. [68Ga]Ga-DOTA-exendin-4 PET/CT proved to be efficacious for longitudinal monitoring of islet transplants. Thus, it could play a crucial role in optimizing ITx as a curative therapy for type 1 diabetes.
Collapse
Affiliation(s)
- Johanna Pilz
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Nicol Gloddek
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Felix Lindheimer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Magdalena J Lindner
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | | | - Muzzafer Ümütlü
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Clemens Cyran
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Max Seidensticker
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Richard Lindner
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Martin Kraetzl
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Simone Renner
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Daphne Merkus
- Walter Brendel Center for Experimental Medicine (WBex), University Hospital, LMU Munich, Munich, Germany
| | - Daniel Teupser
- Department of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Sibylle I Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Elisabeth Kemter
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
4
|
Scholz H, Sordi V, Piemonti L. Cautious Optimism Warranted for Stem Cell-Derived Islet Transplantation in Type 2 Diabetes. Transpl Int 2024; 37:13358. [PMID: 39131791 PMCID: PMC11310020 DOI: 10.3389/ti.2024.13358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/15/2024] [Indexed: 08/13/2024]
Affiliation(s)
- Hanne Scholz
- Department of Transplant Medicine and Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
- Hybrid Technology Hub Centre of Excellence, Institute of Basic Medical Science, University of Oslo, Oslo, Norway
| | - Valeria Sordi
- Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
- Clinic Unit of Regenerative Medicine and Organ Transplants, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
5
|
Marques J, Nunes R, Carvalho AM, Florindo H, Ferreira D, Sarmento B. GLP-1 Analogue-Loaded Glucose-Responsive Nanoparticles as Allies of Stem Cell Therapies for the Treatment of Type I Diabetes. ACS Pharmacol Transl Sci 2024; 7:1650-1663. [PMID: 38751616 PMCID: PMC11092009 DOI: 10.1021/acsptsci.4c00173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/20/2024] [Accepted: 04/24/2024] [Indexed: 05/18/2024]
Abstract
Type 1 diabetes (T1D) is characterized by insufficient insulin secretion due to β-cell loss. Despite exogenous insulin administration being a lifesaving treatment, many patients still experience severe glycemic lability. For these patients, a β-cell replacement strategy through pancreas or pancreatic islet transplantation is the most physiological approach. However, donors' scarcity and the need for lifelong immunosuppressive therapy pose some challenges. This study proposes an innovative biomimetic pancreas, comprising β- and α-cells differentiated from human induced pluripotent stem cells (hiPSCs) embedded in a biofunctional matrix with glucose-responsive nanoparticles (NPs) encapsulating a glucagon-like peptide 1 (GLP-1) analogue, which aims to enhance the glucose responsiveness of differentiated β-cells. Herein, glucose-sensitive pH-responsive NPs encapsulating exenatide or semaglutide showed an average size of 145 nm, with 40% association efficiency for exenatide-loaded NPs and 55% for semaglutide-loaded NPs. Both peptides maintained their secondary structure after in vitro release and showed a similar effect on INS-1E cells' insulin secretion. hiPSCs were differentiated into β- and α-cells, and insulin-positive cells were obtained (82%), despite low glucose responsiveness, as well as glucagon-positive cells (17.5%). The transplantation of the developed system in diabetic mice showed promising outcomes since there was an increase in the survival rate of those animals. Moreover, diabetic mice transplanted with cells and exenatide showed a decrease in their glucose levels. Overall, the biomimetic pancreas developed in this work showed improvements in diabetic mice survival rate, paving the way for new cellular therapies for T1D that explore the synergy of nanomedicines and stem cell-based approaches.
Collapse
Affiliation(s)
- Joana
Moreira Marques
- i3S—Instituto
de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB—Instituto
de Engenharia Biomédica, Universidade
do Porto, Rua Alfredo
Allen, 208, 4200-180 Porto, Portugal
- UCIBIO—Applied
Molecular Biosciences Unit, REQUIMTE, MedTech–Pharmaceutical
Technology Laboratory, Drug Sciences Department, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal
| | - Rute Nunes
- i3S—Instituto
de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IUCS-CESPU
- Instituto Universitário de Ciências da Saúde, 4585-116 Gandra, Portugal
| | - Ana Margarida Carvalho
- i3S—Instituto
de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB—Instituto
de Engenharia Biomédica, Universidade
do Porto, Rua Alfredo
Allen, 208, 4200-180 Porto, Portugal
- ICBAS—Instituto
de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Helena Florindo
- Research
Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Domingos Ferreira
- UCIBIO—Applied
Molecular Biosciences Unit, REQUIMTE, MedTech–Pharmaceutical
Technology Laboratory, Drug Sciences Department, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal
| | - Bruno Sarmento
- i3S—Instituto
de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB—Instituto
de Engenharia Biomédica, Universidade
do Porto, Rua Alfredo
Allen, 208, 4200-180 Porto, Portugal
- IUCS-CESPU
- Instituto Universitário de Ciências da Saúde, 4585-116 Gandra, Portugal
| |
Collapse
|
6
|
O’Brien J, Niehaus P, Chang K, Remark J, Barrett J, Dasgupta A, Adenegan M, Salimian M, Kevas Y, Chandrasekaran K, Kristian T, Chellappan R, Rubin S, Kiemen A, Lu CPJ, Russell JW, Ho CY. Skin keratinocyte-derived SIRT1 and BDNF modulate mechanical allodynia in mouse models of diabetic neuropathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.24.523981. [PMID: 36747753 PMCID: PMC9900813 DOI: 10.1101/2023.01.24.523981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Diabetic neuropathy is a debilitating disorder characterized by spontaneous and mechanical pain. The role of skin mechanoreceptors in the development of mechanical pain (allodynia) is unclear. We discovered that mice with diabetic neuropathy had decreased sirtuin 1 (SIRT1) deacetylase activity in foot skin, leading to reduced expression of brain-derived neurotrophic factor (BDNF) and subsequent loss of innervation in Meissner corpuscles, a mechanoreceptor expressing the BDNF receptor TrkB. When SIRT1 was depleted from skin, the mechanical allodynia worsened in diabetic neuropathy mice, likely due to retrograde degeneration of the Meissner-corpuscle innervating Aβ axons and aberrant formation of Meissner corpuscles which may have increased the mechanosensitivity. The same phenomenon was also noted in skin BDNF knockout mice. Furthermore, overexpression of SIRT1 in skin induced Meissner corpuscle reinnervation and regeneration, resulting in significant improvement of diabetic mechanical allodynia. Overall, the findings suggested that skin-derived SIRT1 and BDNF function in the same pathway in skin sensory apparatus regeneration and highlighted the potential of developing topical SIRT1-activating compounds as a novel treatment for diabetic mechanical allodynia.
Collapse
Affiliation(s)
- Jennifer O’Brien
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Peter Niehaus
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Koping Chang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Pathology, National Taiwan University, Taipei, 100, Taiwan
| | - Juliana Remark
- Hansj rg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Joy Barrett
- Hansj rg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Abhishikta Dasgupta
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Morayo Adenegan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Mohammad Salimian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yanni Kevas
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Krish Chandrasekaran
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD, 21201, USA
| | - Tibor Kristian
- Baltimore Veterans Affairs Medical Center, Baltimore, MD, 21201, USA
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Rajeshwari Chellappan
- Department of Pathology, University of Alabama Birmingham, Birmingham, AL, 35233, USA
| | - Samuel Rubin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Chemistry, College of William and Mary, Williamsburg, VA, 23187, USA
| | - Ashley Kiemen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Catherine Pei-Ju Lu
- Hansj rg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - James W. Russell
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD, 21201, USA
| | - Cheng-Ying Ho
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| |
Collapse
|
7
|
Abadpour S, Niemi EM, Orrhult LS, Hermanns C, de Vries R, Nogueira LP, Haugen HJ, Josefsen D, Krauss S, Gatenholm P, van Apeldoorn A, Scholz H. Adipose-Derived Stromal Cells Preserve Pancreatic Islet Function in a Transplantable 3D Bioprinted Scaffold. Adv Healthc Mater 2023; 12:e2300640. [PMID: 37781993 PMCID: PMC11469278 DOI: 10.1002/adhm.202300640] [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: 02/27/2023] [Revised: 09/07/2023] [Indexed: 10/03/2023]
Abstract
Intra-portal islet transplantation is currently the only clinically approved beta cell replacement therapy, but its outcome is hindered by limited cell survival due to a multifactorial reaction against the allogeneic tissue in liver. Adipose-derived stromal cells (ASCs) can potentially improve the islet micro-environment by their immunomodulatory action. The challenge is to combine both islets and ASCs in a relatively easy and consistent long-term manner in a deliverable scaffold. Manufacturing the 3D bioprinted double-layered scaffolds with primary islets and ASCs using a mix of alginate/nanofibrillated cellulose (NFC) bioink is reported. The diffusion properties of the bioink and the supportive effect of human ASCs on islet viability, glucose sensing, insulin secretion, and reducing the secretion of pro-inflammatory cytokines are demonstrated. Diabetic mice transplanted with islet-ASC scaffolds reach normoglycemia seven days post-transplantation with no significant difference between this group and the group received islets under the kidney capsules. In addition, animals transplanted with islet-ASC scaffolds stay normoglycemic and show elevated levels of C-peptide compared to mice transplanted with islet-only scaffolds. The data present a functional 3D bioprinted scaffold for islets and ASCs transplanted to the extrahepatic site and suggest a possible role of ASCs on improving the islet micro-environment.
Collapse
Affiliation(s)
- Shadab Abadpour
- Department of Transplant MedicineOslo University HospitalOslo0372Norway
- Institute for Surgical ResearchOslo University HospitalOslo0372Norway
- Hybrid Technology Hub – Centre of ExcellenceInstitute of Basic Medical SciencesUniversity of OsloOslo0372Norway
| | - Essi M. Niemi
- Institute for Surgical ResearchOslo University HospitalOslo0372Norway
- Hybrid Technology Hub – Centre of ExcellenceInstitute of Basic Medical SciencesUniversity of OsloOslo0372Norway
- Department of Vascular SurgeryAker HospitalOslo University HospitalOslo0586Norway
| | - Linnea Strid Orrhult
- 3D Bioprinting CenterWWSCDepartment of Chemistry and Chemical EngineeringChalmers University of TechnologyGothenburg41296Sweden
| | - Carolin Hermanns
- MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229The Netherlands
| | - Rick de Vries
- MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229The Netherlands
| | | | | | - Dag Josefsen
- Section for Cellular TherapyRadiumhospitaletOslo University HospitalOslo0379Norway
| | - Stefan Krauss
- Hybrid Technology Hub – Centre of ExcellenceInstitute of Basic Medical SciencesUniversity of OsloOslo0372Norway
- Department of Immunology and Transfusion MedicineOslo University HospitalOslo0372Norway
| | - Paul Gatenholm
- 3D Bioprinting CenterWWSCDepartment of Chemistry and Chemical EngineeringChalmers University of TechnologyGothenburg41296Sweden
- CELLHEAL ASSandvika1337Norway
| | - Aart van Apeldoorn
- MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229The Netherlands
| | - Hanne Scholz
- Department of Transplant MedicineOslo University HospitalOslo0372Norway
- Institute for Surgical ResearchOslo University HospitalOslo0372Norway
- Hybrid Technology Hub – Centre of ExcellenceInstitute of Basic Medical SciencesUniversity of OsloOslo0372Norway
- Section for Cellular TherapyRadiumhospitaletOslo University HospitalOslo0379Norway
| |
Collapse
|
8
|
Krishtul S, Skitel Moshe M, Kovrigina I, Baruch L, Machluf M. ECM-based bioactive microencapsulation significantly improves islet function and graft performance. Acta Biomater 2023; 171:249-260. [PMID: 37708927 DOI: 10.1016/j.actbio.2023.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 08/17/2023] [Accepted: 09/07/2023] [Indexed: 09/16/2023]
Abstract
Microencapsulation is a promising strategy to prolong the survival and function of transplanted pancreatic islets for diabetes therapy, albeit its translation has been impeded by incoherent graft performance. The use of decellularized ECM has lately gained substantial research momentum due to its innate capacity to augment the function of cells originating from the same tissue type. In the present study, the advantages of both these approaches are leveraged in a porcine pancreatic ECM (pECM)-based microencapsulation platform, thus significantly enhancing murine pancreatic islet performance. pECM-encapsulated islets sustain high insulin secretion levels in vitro, surpassing those of islets encapsulated in conventional alginate microcapsules. Moreover, pECM-encapsulated islet cells proliferate and produce an enriched intra-islet ECM framework, displaying a distinctive structural rearrangement. The beneficial effect of pECM encapsulation is further reinforced by the temporary protection against cytokine-induced cytotoxicity. In-vivo, this platform significantly improves glucose tolerance and achieves glycemic correction in 100% of immunocompetent diabetic mice without any immunosuppression, compared to only 50% mice achieved glycemic correction by alginate encapsulation. Altogether, the results presented herein reveal that pECM-based microencapsulation offers a natural pancreatic niche that can restore the function of isolated pancreatic islets and deliver them safely, avoiding the need for immunosuppression. STATEMENT OF SIGNIFICANCE: Aiming to improve pancreatic islet transplantation outcomes in diabetic patients, we developed a microencapsulation platform based on pancreatic extracellular matrix (pECM). In these microcapsules the islets are entrapped within a pECM hydrogel that mimics the natural pancreatic microenvironment. We show that pECM encapsulation supports the islets' viability and function in culture, and provides temporal protection against cytokine-induced stress. In a diabetic mouse model, pECM encapsulation significantly improved glucose tolerance and achieved glycemic correction without any immunosuppression. These results reveal the potential of pECM encapsulation as a viable treatment for diabetes, providing a solid scientific basis for more advanced preclinical studies.
Collapse
Affiliation(s)
- Stasia Krishtul
- Faculty of Biotechnology & Food Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Michal Skitel Moshe
- Faculty of Biotechnology & Food Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Inna Kovrigina
- Faculty of Biotechnology & Food Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Limor Baruch
- Faculty of Biotechnology & Food Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Marcelle Machluf
- Faculty of Biotechnology & Food Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel.
| |
Collapse
|
9
|
Ernst AU, Wang LH, Worland SC, Marfil-Garza BA, Wang X, Liu W, Chiu A, Kin T, O'Gorman D, Steinschneider S, Datta AK, Papas KK, James Shapiro AM, Ma M. A predictive computational platform for optimizing the design of bioartificial pancreas devices. Nat Commun 2022; 13:6031. [PMID: 36229614 PMCID: PMC9561707 DOI: 10.1038/s41467-022-33760-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 09/30/2022] [Indexed: 11/09/2022] Open
Abstract
The delivery of encapsulated islets or stem cell-derived insulin-producing cells (i.e., bioartificial pancreas devices) may achieve a functional cure for type 1 diabetes, but their efficacy is limited by mass transport constraints. Modeling such constraints is thus desirable, but previous efforts invoke simplifications which limit the utility of their insights. Herein, we present a computational platform for investigating the therapeutic capacity of generic and user-programmable bioartificial pancreas devices, which accounts for highly influential stochastic properties including the size distribution and random localization of the cells. We first apply the platform in a study which finds that endogenous islet size distribution variance significantly influences device potency. Then we pursue optimizations, determining ideal device structures and estimates of the curative cell dose. Finally, we propose a new, device-specific islet equivalence conversion table, and develop a surrogate machine learning model, hosted on a web application, to rapidly produce these coefficients for user-defined devices.
Collapse
Affiliation(s)
- Alexander U Ernst
- Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Long-Hai Wang
- Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA.
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China.
| | - Scott C Worland
- Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | | | - Xi Wang
- Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Wanjun Liu
- Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Alan Chiu
- Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Tatsuya Kin
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Doug O'Gorman
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | | | - Ashim K Datta
- Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | | | - A M James Shapiro
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Minglin Ma
- Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
10
|
Porret R, Meier RPH, Mikulic J, Pascual M, Aubert V, Harr T, Golshayan D, Muller YD. Atopy as an independent predictor for long-term patient and graft survival after kidney transplantation. Front Immunol 2022; 13:997364. [PMID: 36263051 PMCID: PMC9574189 DOI: 10.3389/fimmu.2022.997364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundAtopy is a genetic condition predisposing individuals to develop immunoglobulin E (IgE) against common allergens through T-helper 2 (Th2) polarization mechanisms. The impact of atopy on graft survival in solid organ transplantation is unknown.MethodologyWe analyzed 268 renal allograft recipients from the Swiss Transplant Cohort Study, a prospective multicenter cohort studying patients after solid organ transplantation, with a 9-year median follow-up (IQR 3.0). We used the Phadiatop assay to measure IgE antibodies against a mixture of common inhaled allergens (grass, tree, herbs, spores, animals, and mites) to identify pre-transplantation atopic patients (>0.35 KU/L).ResultsOf 268 kidney transplant recipients, 66 individuals were atopic (24.6%). Atopic patients were significantly younger than non-atopic patients (49.6 vs 58.0 years old, P = 0.002). No significant difference was found for gender, cold/warm ischemia time, preformed donor-specific antibodies (DSA), HLA mismatches, induction and maintenance immunosuppressive therapy, CMV serostatus, or cause of kidney failure. Patient and graft survival at ten years of follow-up were significantly better in the atopic group, 95.2% versus 69.2% patient survival (P < 0.001), and 87.9% versus 60.8% graft survival (P < 0.001), respectively. A multivariate Cox analysis revealed that atopy predicted recipient and graft survival independently of age and living donor donation. Finally, we found similar rates of biopsy-proven acute cellular and antibody-mediated rejections between atopic and non-atopic recipients.ConclusionAtopy was associated with better long-term patient and graft survival, independently of age and living donor donation after kidney transplantation. Yet, atopy should not be used as a predictor for acute rejection.
Collapse
Affiliation(s)
- Raphaël Porret
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Raphaël P. H. Meier
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Josip Mikulic
- Transplantation Center, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Manuel Pascual
- Transplantation Center, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Vincent Aubert
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Thomas Harr
- Division of Immunology and Allergy, University Hospitals of Geneva, Geneva, Switzerland
| | - Déla Golshayan
- Transplantation Center, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Yannick D. Muller
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
- *Correspondence: Yannick D. Muller,
| |
Collapse
|
11
|
Moazenchi M, Sadr Hashemi Nejad A, Izadi M, Khalaj M, Samsonchi Z, Tavakol Rad P, Amini P, Tahamtani Y, Hajizadeh-Saffar E. Comparative Study of The Effects of Confounding Factors on Improving Rat Pancreatic Islet Isolation Yield and Quality. CELL JOURNAL 2022; 24:491-499. [PMID: 36274201 PMCID: PMC9588161 DOI: 10.22074/cellj.2022.8123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Isolated pancreatic islets are valuable resources for a wide range of research, including cell replacement studies and cell-based platforms for diabetes drug discovery and disease modeling. Islet isolation is a complex and stepwise procedure aiming to obtain pure, viable, and functional islets for in vitro and in vivo studies. It should be noted that differences in rodent strains, gender, weight, and density gradients may affect the isolated islet's properties. We evaluated the variables affecting the rat islet isolation procedure to reach the maximum islet yield and functionality, which would be critical for further studies on islet regenerative biology. MATERIALS AND METHODS The present experimental study compared the yield and purity of isolated islets from nondiabetic rats of two different strains. Next, islet particle number (IPN) and islet equivalent (IEQ) were compared between males and females, and the weight range that yields the highest number of islets was investigated. Moreover, the influence of three different density gradients, namely Histopaque, Pancoll, and Lymphodex, on final isolated islets purity and yield were assessed. Finally, the viability and functionality of isolated islets were measured. RESULTS The IEQ, IPN, and purity of isolated islets in 15 Lister hooded rats (LHRs) were significantly (P≤0.05) higher than those of the other strains. Male LHRs resulted in significantly higher IEQ compared to females (P≤0.05). Moreover, IPN and IEQ did not significantly vary among different weight groups. Also, the utilization of Histopaque and Pancoll leads to higher yield and purity. In vivo assessments of the isolated islets presented significantly reduced blood glucose percentage in the transplanted group on days 2-5 following transplantation. CONCLUSION Based on these results, an optimal protocol for isolating high-quality rat islets with a constant yield, purity, and function has been established as an essential platform for developing diabetes research.
Collapse
Affiliation(s)
- Maedeh Moazenchi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran,Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute
for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Anavasadat Sadr Hashemi Nejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran,Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute
for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahmoud Izadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran,Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute
for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maedeh Khalaj
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran
| | - Zakieh Samsonchi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran
| | - Pouya Tavakol Rad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran
| | - Payam Amini
- Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Yaser Tahamtani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran,Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Ensiyeh Hajizadeh-Saffar
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute
for Stem Cell Biology and Technology, ACECR, Tehran, Iran,Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran,Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR,
Tehran, Iran,P.O.Box: 16635-148Department of Regenerative MedicineCell Science Research CenterRoyan Institute for Stem Cell
Biology and TechnologyACECRTehranIran
| |
Collapse
|
12
|
Zhang L, Turan A, Shirwan H, Yolcu ES. A modified surgical procedure using minimally invasive ileocolic vein perfusion in a mouse intrahepatic islet transplant model. STAR Protoc 2022; 3:101416. [PMID: 35620067 PMCID: PMC9127695 DOI: 10.1016/j.xpro.2022.101416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Murine intrahepatic islet transplantation is a clinically relevant but technically challenging surgical procedure because of frequent lethal postoperative bleeding. Here, we describe a protocol for mouse pancreatic islet isolation, purification, and culture. Besides, we also describe a protocol for intrahepatic islet transplantation through the ileocolic vein. Intrahepatic islet transplantation through the ileocolic vein, as opposed to traditional islet perfusion via the main portal vein, has the advantage of improving recovery after surgery and may facilitate islet survival and function in preclinical settings. For complete details on the use and execution of this protocol, please refer to Shrestha et al. (2020).
Collapse
Affiliation(s)
- Lei Zhang
- Department of Child Health, University of Missouri, Columbia, MO 65211, USA
| | - Ali Turan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO 65211, USA
| | - Haval Shirwan
- Department of Child Health, University of Missouri, Columbia, MO 65211, USA
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO 65211, USA
| | - Esma S. Yolcu
- Department of Child Health, University of Missouri, Columbia, MO 65211, USA
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
13
|
Burke JA, Zhang X, Bobbala S, Frey MA, Bohorquez Fuentes C, Freire Haddad H, Allen SD, Richardson RAK, Ameer GA, Scott EA. Subcutaneous nanotherapy repurposes the immunosuppressive mechanism of rapamycin to enhance allogeneic islet graft viability. NATURE NANOTECHNOLOGY 2022; 17:319-330. [PMID: 35039683 PMCID: PMC8934301 DOI: 10.1038/s41565-021-01048-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/09/2021] [Indexed: 05/03/2023]
Abstract
Standard oral rapamycin (that is, Rapamune) administration is plagued by poor bioavailability and broad biodistribution. Thus, this pleotropic mammalian target of rapamycin (mTOR) inhibitor has a narrow therapeutic window and numerous side effects and provides inadequate protection to transplanted cells and tissues. Furthermore, the hydrophobicity of rapamycin limits its use in parenteral formulations. Here, we demonstrate that subcutaneous delivery via poly(ethylene glycol)-b-poly(propylene sulfide) polymersome nanocarriers significantly alters rapamycin's cellular biodistribution to repurpose its mechanism of action for tolerance, instead of immunosuppression, and minimize side effects. While oral rapamycin inhibits T cell proliferation directly, subcutaneously administered rapamycin-loaded polymersomes modulate antigen presenting cells in lieu of T cells, significantly improving maintenance of normoglycemia in a clinically relevant, major histocompatibility complex-mismatched, allogeneic, intraportal (liver) islet transplantation model. These results demonstrate the ability of a rationally designed nanocarrier to re-engineer the immunosuppressive mechanism of a drug by controlling cellular biodistribution.
Collapse
Affiliation(s)
- Jacqueline A Burke
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Xiaomin Zhang
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Molly A Frey
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA
| | - Carolina Bohorquez Fuentes
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Helena Freire Haddad
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Sean D Allen
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA
| | - Reese A K Richardson
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA
| | - Guillermo A Ameer
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA.
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA.
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA.
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA.
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA.
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA.
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA.
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA.
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA.
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
14
|
Kattner AA. The best protection is early detection. Biomed J 2022; 44:S155-S161. [PMID: 35042015 PMCID: PMC9068561 DOI: 10.1016/j.bj.2022.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 10/29/2022] Open
Abstract
This current special issue of the Biomedical Journal provides insights in various cancer forms, and possible ways of prognostic and predictive screening. In detail we learn about lung cancer and tissue samples from ground glass opacifications, liquid biopsy through circulating tumor cells in colon cancer, transcription factor analysis in cervical cancer, and long non-coding RNAs in breast and lung cancer. A prognosis factor in individuals with acute myeloid leukemia and a rare fungal infection are determined. Challenges surrounding transplantation are elucidated, a potential biomarker for allograft dysfunction is presented, as well as a mean to save beta cells after islet transplantation. We get to know more about drug resistance in transplant recipients with tuberculosis, and also in the case of H.pylori infection. Lastly, the possibilities of cardiac shock wave therapy in simultaneous artery and renal disease is explored, we are presented with genetic factors contributing to cancer risk in arseniasis areas, and protocol recommendations for the optimal reproducibility of bladder volume in prostate cancer treatment. Three markers for detecting stages of diabetic retinopathy are covered, as well as a way to mitigate effects of lungworm secretions. Finally we get to see a novel approach for acupuncture needle material, and two management approaches for a form of skeletal malocclusion.
Collapse
|
15
|
Current Status, Barriers, and Future Directions for Humanized Mouse Models to Evaluate Stem Cell–Based Islet Cell Transplant. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1387:89-106. [DOI: 10.1007/5584_2022_711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
16
|
Streptozotocin-Induced Diabetes in a Mouse Model (BALB/c) Is Not an Effective Model for Research on Transplantation Procedures in the Treatment of Type 1 Diabetes. Biomedicines 2021; 9:biomedicines9121790. [PMID: 34944607 PMCID: PMC8698562 DOI: 10.3390/biomedicines9121790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/14/2022] Open
Abstract
Type 1 diabetes (T1D) is characterized by the destruction of over 90% of the β-cells. C-peptide is a parameter for evaluating T1D. Streptozotocin (STZ) is a standard method of inducing diabetes in animals. Eight protocols describe the administration of STZ in mice; C-peptide levels are not taken into account. The aim of the study is to determine whether the STZ protocol for the induction of beta-cell mass destruction allows for the development of a stable in vivo mouse model for research into new transplant procedures in the treatment of type 1 diabetes. Materials and methods: Forty BALB/c mice were used. The animals were divided into nine groups according to the STZ dose and a control group. The STZ doses were between 140 and 400 mg/kg of body weight. C-peptide was taken before and 2, 7, 9, 12, 14, and 21 days after STZ. Immunohistochemistry was performed. The area of the islet and insulin-/glucagon-expressing tissues was calculated. Results: Mice who received 140, 160, 2 × 100, 200, and 250 mg of STZ did not show changes in mean fasting C-peptide in comparison to the control group and to day 0. All animals with doses of 300 and 400 mg of STZ died during the experiment. The area of the islets did not show any differences between the control and STZ-treated mice in groups below 300 mg. The reduction of insulin-positive areas in STZ mice did not exceed 50%. Conclusions: Streptozotocin is not an appropriate method of inducing a diabetes model for further research on transplantation treatments of type 1 diabetes, having caused the destruction of more than 90% of the β-cell mass in BALB/c mice.
Collapse
|
17
|
Razavi M, Wang J, Thakor AS. Localized drug delivery graphene bioscaffolds for cotransplantation of islets and mesenchymal stem cells. SCIENCE ADVANCES 2021; 7:eabf9221. [PMID: 34788097 PMCID: PMC8597999 DOI: 10.1126/sciadv.abf9221] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 09/28/2021] [Indexed: 06/01/2023]
Abstract
In the present work, we developed, characterized, and tested an implantable graphene bioscaffold which elutes dexamethasone (Dex) that can accommodate islets and adipose tissue–derived mesenchymal stem cells (AD-MSCs). In vitro studies demonstrated that islets in graphene–0.5 w/v% Dex bioscaffolds had a substantial higher viability and function compared to islets in graphene-alone bioscaffolds or islets cultured alone (P < 0.05). In vivo studies, in which bioscaffolds were transplanted into the epididymal fat pad of diabetic mice, demonstrated that, when islet:AD-MSC units were seeded into graphene–0.5 w/v% Dex bioscaffolds, this resulted in complete restoration of glycemic control immediately after transplantation with these islets also showing a faster response to glucose challenges (P < 0.05). Hence, this combination approach of using a graphene bioscaffold that can be functionalized for local delivery of Dex into the surrounding microenvironment, together with AD-MSC therapy, can significantly improve the survival and function of transplanted islets.
Collapse
Affiliation(s)
- Mehdi Razavi
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Biionix™ (Bionic Materials, Implants & Interfaces) Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA
| | - Jing Wang
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Avnesh S. Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| |
Collapse
|
18
|
Zamboni F, Cengiz IF, Barbosa AM, Castro AG, Reis RL, Oliveira JM, Collins MN. Towards the Development of a Female Animal Model of T1DM Using Hyaluronic Acid Nanocoated Cell Transplantation: Refinements and Considerations for Future Protocols. Pharmaceutics 2021; 13:pharmaceutics13111925. [PMID: 34834340 PMCID: PMC8621706 DOI: 10.3390/pharmaceutics13111925] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/03/2021] [Accepted: 11/10/2021] [Indexed: 12/11/2022] Open
Abstract
Female mice (Black 6 strain) (C57BL/6) aged 6 weeks were subject to low dose streptozotocin (STZ) treatment for five consecutive days to mimic type 1 diabetes mellitus (T1DM) with insulitis. At two weeks after STZ injections, evaluation of the elevated glucose levels was used to confirm diabetes. The diabetic mice were then subject to the transplantation of pancreatic β-cells (MIN-6 line). Four groups of mice were studied. The first group was injected with saline-only acting as the placebo surgery control, also known as SHAM group, the second and third groups were injected with MIN-6 single cells and polyethylene glycol-modified dipalmitoyl-glycerol-phosphatidyl ethanolamine (PEG-DPPE) modified MIN-6 single cells (500 µg per 1.106 cells), respectively, while the fourth group was injected with hyaluronic acid (HA)-coated MIN-6 single cells (5 bilayers). At seven- and fourteen-days following transplantation, the mice were euthanised. The renal and pancreatic tissues were then collected and histologically analysed. The induction of diabetes in female mice, through five-consecutive daily STZ injections resulted in inconsistent glycaemic levels. Interestingly, this shows an incomplete diabetes induction in female mice, of which we attribute to sex dimorphism and hormonal interferences. Transplantation failure of free-floating encapsulated cells was unable to decrease blood glucose hyperglycaemia to physiological ranges. The result is attributed to deprived cell–cell interactions, leading to decreased β-cells functionality. Overall, we highlight the necessity of refining T1DM disease models in female subjects when using multiple low-dose STZ injections together with transplantation protocols. Considerations need to be made regarding the different developmental stages of female mice and oestrogen load interfering with pancreatic β-cells susceptibility to STZ. The use of pseudo islets, cell aggregates and spheroids are sought to improve transplantation outcome in comparison to free-floating single cells.
Collapse
Affiliation(s)
- Fernanda Zamboni
- Stokes Laboratories, School of Engineering, Bernal Institute, University of Limerick, Limerick V94 T9PX, Ireland;
- Health Research Institute, University of Limerick, Limerick V94 T9PX, Ireland
| | - Ibrahim F. Cengiz
- 13B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; (I.F.C.); (R.L.R.); (J.M.O.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Braga, Portugal; (A.M.B.); (A.G.C.)
| | - Ana M. Barbosa
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Braga, Portugal; (A.M.B.); (A.G.C.)
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Braga, Portugal
| | - Antonio G. Castro
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Braga, Portugal; (A.M.B.); (A.G.C.)
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Braga, Portugal
| | - Rui L. Reis
- 13B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; (I.F.C.); (R.L.R.); (J.M.O.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Braga, Portugal; (A.M.B.); (A.G.C.)
| | - Joaquim M. Oliveira
- 13B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; (I.F.C.); (R.L.R.); (J.M.O.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Braga, Portugal; (A.M.B.); (A.G.C.)
| | - Maurice N. Collins
- Stokes Laboratories, School of Engineering, Bernal Institute, University of Limerick, Limerick V94 T9PX, Ireland;
- Health Research Institute, University of Limerick, Limerick V94 T9PX, Ireland
- SFI AMBER, University of Limerick, Limerick V94 T9PX, Ireland
- Correspondence:
| |
Collapse
|
19
|
Abstract
This review focuses on the human pancreatic islet-including its structure, cell composition, development, function, and dysfunction. After providing a historical timeline of key discoveries about human islets over the past century, we describe new research approaches and technologies that are being used to study human islets and how these are providing insight into human islet physiology and pathophysiology. We also describe changes or adaptations in human islets in response to physiologic challenges such as pregnancy, aging, and insulin resistance and discuss islet changes in human diabetes of many forms. We outline current and future interventions being developed to protect, restore, or replace human islets. The review also highlights unresolved questions about human islets and proposes areas where additional research on human islets is needed.
Collapse
Affiliation(s)
- John T Walker
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Diane C Saunders
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Marcela Brissova
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Alvin C Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
20
|
Forbes S, Bond AR, Thirlwell KL, Burgoyne P, Samuel K, Noble J, Borthwick G, Colligan D, McGowan NWA, Lewis PS, Fraser AR, Mountford JC, Carter RN, Morton NM, Turner ML, Graham GJ, Campbell JDM. Human umbilical cord perivascular cells improve human pancreatic islet transplant function by increasing vascularization. Sci Transl Med 2021; 12:12/526/eaan5907. [PMID: 31941825 DOI: 10.1126/scitranslmed.aan5907] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 06/24/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022]
Abstract
Islet transplantation is an efficacious therapy for type 1 diabetes; however, islets from multiple donor pancreata are required, and a gradual attrition in transplant function is seen. Here, we manufactured human umbilical cord perivascular mesenchymal stromal cells (HUCPVCs) to Good Manufacturing Practice (GMP) standards. HUCPVCs showed a stable phenotype while undergoing rapid ex vivo expansion at passage 2 (p2) to passage 4 (p4) and produced proregenerative factors, strongly suppressing T cell responses in the resting state and in response to inflammation. Transplanting an islet equivalent (IEQ):HUCPVC ratio of 1:30 under the kidney capsule in diabetic NSG mice demonstrated the fastest return to normoglycemia by 3 days after transplant: Superior glycemic control was seen at both early (2.7 weeks) and later stages (7, 12, and 16 weeks) versus ratios of 1:0, 1:10, and 1:50, respectively. Syngeneic islet transplantation in immunocompetent mice using the clinically relevant hepatic portal route with a marginal islet mass showed that mice transplanted with an IEQ:HUCPVC ratio of 1:150 had superior glycemic control versus ratios of 1:0, 1:90, and 1:210 up to 6 weeks after transplant. Immunodeficient mice transplanted with human islets (IEQ:HUCPVC ratio of 1:150) exhibited better glycemic control for 7 weeks after transplant versus islet transplant alone, and islets transplanted via the hepatic portal vein in an allogeneic mouse model using a curative islet mass demonstrated delayed rejection of islets when cotransplanted with HUCPVCs (IEQ:HUCPVC ratio of 1:150). The immunosuppressive and proregenerative properties of HUCPVCs demonstrated long-term positive effects on graft function in vivo, indicating that they may improve long-term human islet allotransplantation outcomes.
Collapse
Affiliation(s)
- Shareen Forbes
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK. .,Clinical Islet Transplantation Programme, Royal Infirmary of Edinburgh, Edinburgh EH16 4SU, UK
| | - Andrew R Bond
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Kayleigh L Thirlwell
- Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh EH14 4BE, UK.,Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| | - Paul Burgoyne
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK.,Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh EH14 4BE, UK.,Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| | - Kay Samuel
- Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh EH14 4BE, UK
| | - June Noble
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Gary Borthwick
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - David Colligan
- Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh EH14 4BE, UK
| | - Neil W A McGowan
- Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh EH14 4BE, UK
| | - Philip Starkey Lewis
- Medical Research Council (MRC) Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Alasdair R Fraser
- Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh EH14 4BE, UK
| | - Joanne C Mountford
- Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh EH14 4BE, UK
| | - Roderick N Carter
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Nicholas M Morton
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Marc L Turner
- Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh EH14 4BE, UK
| | - Gerard J Graham
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| | - John D M Campbell
- Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh EH14 4BE, UK. .,Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| |
Collapse
|
21
|
Primavera R, Razavi M, Kevadiya BD, Wang J, Vykunta A, Di Mascolo D, Decuzzi P, Thakor AS. Enhancing islet transplantation using a biocompatible collagen-PDMS bioscaffold enriched with dexamethasone-microplates. Biofabrication 2021; 13. [PMID: 33455953 DOI: 10.1088/1758-5090/abdcac] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/15/2021] [Indexed: 01/01/2023]
Abstract
Islet transplantation is a promising approach to enable type 1 diabetic patients to attain glycemic control independent of insulin injections. However, up to 60% of islets are lost immediately following transplantation. To improve this outcome, islets can be transplanted within bioscaffolds, however, synthetic bioscaffolds induce an intense inflammatory reaction which can have detrimental effects on islet function and survival. In the present study, we first improved the biocompatibility of polydimethylsiloxane (PDMS) bioscaffolds by coating them with collagen. To reduce the inflammatory response to PDMS bioscaffolds, we then enriched the bioscaffolds with dexamethasone-loaded microplates (DEX-µScaffolds). These DEX-microplates have the ability to release DEX in a sustained manner over 7 weeks within a therapeutic range that does not affect the glucose responsiveness of the islets but which minimizes inflammation in the surrounding microenvironment. The bioscaffold showed excellent mechanical properties that enabled it to resist pore collapse thereby helping to facilitate islet seeding and its handling for implantation, and subsequent engraftment, within the epididymal fat pad (EFP). Following the transplantation of islets into the EFP of diabetic mice using DEX-µScaffolds there was a return in basal blood glucose to normal values by day 4, with normoglycemia maintained for 30 days. Furthermore, these animals demonstrated a normal dynamic response to glucose challenges with histological evidence showing reduced pro-inflammatory cytokines and fibrotic tissue surrounding DEX-µScaffolds at the transplantation site. In contrast, diabetic animals transplanted with either islets alone or islets in bioscaffolds without DEX microplates were not able to regain glycemic control during basal conditions with overall poor islet function. Taken together, our data show that coating PDMS bioscaffolds with collagen, and enriching them with DEX-microplates, significantly prolongs and enhances islet function and survival.
Collapse
Affiliation(s)
- Rosita Primavera
- Radiology, Stanford University School of Medicine, 3155 Porter Drive, Stanford, California, 94305-5119, UNITED STATES
| | - Mehdi Razavi
- University of Central Florida, 6900 Lake Nona Blvd, Orlando, Florida, 32827, UNITED STATES
| | - Bhavesh D Kevadiya
- PEN, University of Nebraska Medical Center, Lab-3064,DRC-1,department of pharmacology and experimental neuroscience, Omaha, Nebraska, 68198, UNITED STATES
| | - Jing Wang
- Radiology, Stanford University School of Medicine, 3155 Porter Drive, Stanford, California, 94304, UNITED STATES
| | - Akshara Vykunta
- Radiology, Stanford University School of Medicine, 3155 Porter Drive, Stanford, California, 94304, UNITED STATES
| | - Daniele Di Mascolo
- Central Research Labs Genova, Istituto Italiano di Tecnologia, Via Morego, 30, Genova, Liguria, 16163, ITALY
| | - Paolo Decuzzi
- Istituto Italiano di Tecnologia, Via Morego, 30, Genova, Liguria, 16163, ITALY
| | - Avnesh S Thakor
- Radiology, Stanford University School of Medicine, 3155 Porter Drive, Stanford, California, 94304, UNITED STATES
| |
Collapse
|
22
|
Mridha AR, Dargaville TR, Dalton PD, Carroll L, Morris MB, Vaithilingam V, Tuch BE. Prevascularized Retrievable Hybrid Implant to Enhance Function of Subcutaneous Encapsulated Islets. Tissue Eng Part A 2020; 28:212-224. [PMID: 33081600 DOI: 10.1089/ten.tea.2020.0179] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Replacement of pancreatic β-cells is one of the most promising treatment options for treatment of type 1 diabetes (T1D), even though, toxic immunosuppressive drugs are required. In this study, we aim to deliver allogeneic β-cell therapies without antirejection drugs using a bioengineered hybrid device that contains microencapsulated β-cells inside 3D polycaprolactone (PCL) scaffolds printed using melt electrospin writing (MEW). Mouse β-cell (MIN6) pseudoislets and QS mouse islets are encapsulated in alginate microcapsules, without affecting viability and insulin secretion. Microencapsulated MIN6 cells are then seeded within 3D MEW scaffolds, and these hybrid devices implanted subcutaneously in streptozotocin-treated diabetic NOD/SCID and BALB/c mice. Similar to NOD/SCID mice, blood glucose levels (BGL) are lowered from 30.1 to 4.8 mM in 25-41 days in BALB/c. In contrast, microencapsulated islets placed in prevascularized MEW scaffold 3 weeks after implantation in BALB/c mice normalize BGL (<12 mM) more rapidly, lasting for 60-105 days. The lowering of glucose levels is confirmed by an intraperitoneal glucose tolerance test. Vascularity within the implanted grafts is demonstrated and quantified by 3D-doppler ultrasound, with a linear increase over 4 weeks (r = 0.65). Examination of the device at 5 weeks shows inflammatory infiltrates of neutrophils, macrophages, and B-lymphocytes on the MEW scaffolds, but not on microcapsules, which have infrequent profibrotic walling. In conclusion, we demonstrate the fabrication of an implantable and retrievable hybrid device for vascularization and enhancing the survival of encapsulated islets implanted subcutaneously in an allotransplantation setting without immunosuppression. This study provides proof-of-concept for the application of such devices for human use, but, will require modifications to allow translation to people with T1D. Impact statement The retrievable 3D printed PCL scaffold we have produced promotes vascularization when implanted subcutaneously and allows seeded microencapsulated insulin-producing cells to normalize blood glucose of diabetic mice for at least 2 months, without the need for antirejection drugs to be administered. The scaffold is scalable for possible human use, but will require modification to ensure that normalization of blood glucose levels can be maintained long term.
Collapse
Affiliation(s)
- Auvro R Mridha
- Discipline of Physiology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, Australia.,Australian Foundation for Diabetes Research, Sydney, Australia.,Bosch Institute, The University of Sydney, Sydney, Australia
| | - Tim R Dargaville
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Queensland, Australia
| | - Paul D Dalton
- Department of Functional Materials in Medicine and Dentistry, University of Würzburg, Würzburg, Germany
| | - Luke Carroll
- Discipline of Physiology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, Australia.,Australian Foundation for Diabetes Research, Sydney, Australia.,Now Based at NHMRC Clinical Trials Centre, The University of Sydney, Sydney, Australia
| | - Michael B Morris
- Discipline of Physiology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, Australia.,Bosch Institute, The University of Sydney, Sydney, Australia
| | - Vijayaganapathy Vaithilingam
- Australian Foundation for Diabetes Research, Sydney, Australia.,Cell Biology Inspired Tissue Engineering (CBITE), MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Bernard E Tuch
- Discipline of Physiology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, Australia.,Australian Foundation for Diabetes Research, Sydney, Australia
| |
Collapse
|
23
|
A scalable device-less biomaterial approach for subcutaneous islet transplantation. Biomaterials 2020; 269:120499. [PMID: 33168223 DOI: 10.1016/j.biomaterials.2020.120499] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/01/2020] [Accepted: 10/26/2020] [Indexed: 01/09/2023]
Abstract
The subcutaneous space has been shown to be a suitable site for islet transplantation, however an abundance of islets is required to achieve normoglycemia, often requiring multiple donors. The loss of islets is due to the hypoxic conditions islets experience during revascularization, resulting in apoptosis. Therefore, to reduce the therapeutic dosage required to achieve normoglycemia, pre-vascularization of the subcutaneous space has been pursued. In this study, we highlight a biomaterial-based approach using a methacrylic acid copolymer coating to generate a robust pre-vascularized subcutaneous cavity for islet transplantation. We also devised a simple, but not-trivial, procedure for filling the cavity with an islet suspension in collagen. We show that the pre-vascularized site can support a marginal mass of islets to rapidly return streptozotocin-induced diabetic SCID/bg mice to normoglycemia. Furthermore, immunocompetent Sprague Daley rats remained normoglycemia for up to 70 days until they experienced graft destabilization as they outgrew their implants. This work highlights methacrylic acid-based biomaterials as a suitable pre-vascularization strategy for the subcutaneous space that is scalable and doesn't require exogenous cells or growth factors.
Collapse
|
24
|
Yamane K, Anazawa T, Tada S, Fujimoto N, Inoguchi K, Emoto N, Nagai K, Masui T, Okajima H, Takaori K, Sumi S, Uemoto S. Mitomycin C treatment improves pancreatic islet graft longevity in intraportal islet transplantation by suppressing proinflammatory response. Sci Rep 2020; 10:12086. [PMID: 32694579 PMCID: PMC7374693 DOI: 10.1038/s41598-020-69009-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 07/06/2020] [Indexed: 12/14/2022] Open
Abstract
The in vitro culture period prior to cell transplantation (i.e. pancreatic islet transplantation) enables cell modification and is thus advantageous. However, the islet preconditioning method has not been fully explored. Here we present a simple approach for islet preconditioning that uses the antibiotic mitomycin C (MMC), which has antitumor activity, to reduce islet immunogenicity and prevent proinflammatory events in an intraportal islet transplantation model. Freshly isolated mice islets were treated for 30 min with 10 μg/mL MMC or not, cultured for 20 h and transplanted into the livers of syngeneic or allogeneic diabetic mouse recipients. In the allogeneic model, MMC preconditioning significantly prolonged graft survival without requiring immunosuppressants. In vitro, MMC treatment suppressed the expression of proinflammatory cytokines in islet allografts, while immunohistochemical studies revealed the suppression of inflammatory cell infiltration into MMC-treated allografts relative to untreated allografts. Furthermore, MMC preconditioning significantly suppressed the mRNA expression of proinflammatory cytokines into the transplant site and induced the differentiation of regulatory T cells with the ability to suppress CD4+ T cell-mediated immune responses. In conclusion, islet preconditioning with MMC prolonged graft survival in an intraportal islet transplantation model by suppressing proinflammatory events and inducing potentially regulatory lymphocytes.
Collapse
Affiliation(s)
- Kei Yamane
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Takayuki Anazawa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan.
| | - Seiichiro Tada
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Nanae Fujimoto
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Kenta Inoguchi
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Norio Emoto
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Kazuyuki Nagai
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Toshihiko Masui
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Hideaki Okajima
- Department of Paediatric Surgery, Kanazawa Medical University, Kanazawa, 9200293, Japan
| | - Kyoichi Takaori
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Shoichiro Sumi
- Laboratory of Organ and Tissue Reconstruction, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, 6068507, Japan
| | - Shinji Uemoto
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| |
Collapse
|
25
|
Delcassian D, Luzhansky I, Spanoudaki V, Bochenek M, McGladrigan C, Nguyen A, Norcross S, Zhu Y, Shan CS, Hausser R, Shakesheff KM, Langer R, Anderson DG. Magnetic Retrieval of Encapsulated Beta Cell Transplants from Diabetic Mice Using Dual-Function MRI Visible and Retrievable Microcapsules. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1904502. [PMID: 32134138 DOI: 10.1002/adma.201904502] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 12/03/2019] [Indexed: 05/18/2023]
Abstract
Encapsulated beta cell transplantation offers a potential cure for a subset of diabetic patients. Once transplanted, beta cell grafts can help to restore glycemic control; however, locating and retrieving cells in the event of graft failure may pose a surgical challenge. Here, a dual-function nanoparticle-loaded hydrogel microcapsule is developed that enables graft retrieval under an applied magnetic field. Additionally, this system facilitates graft localization via magnetic resonance imaging (MRI), and graft isolation from the immune system. Iron oxide nanoparticles encapsulated within alginate hydrogel capsules containing viable islets are transplanted and the in vitro and in vivo retrieval of capsules containing nanoparticles functionalized with various ligands are compared. Capsules containing islets co-encapsulated with COOH-coated nanoparticles restore normal glycemia in immunocompetent diabetic mice for at least 6 weeks, can be visualized using MRI, and are retrievable in a magnetic field. Application of a magnetic field for 90 s via a magnetically assisted retrieval device facilitates rapid retrieval of up to 94% (±3.1%) of the transplant volume 24 h after surgical implantation. This strategy aids monitoring of cell-capsule locations in vivo, facilitates graft removal at the end of the transplant lifetime, and may be applicable to many encapsulated cell transplant systems.
Collapse
Affiliation(s)
- Derfogail Delcassian
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Division of Regenerative Medicine and Cellular Therapies, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Igor Luzhansky
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Virginia Spanoudaki
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Matthew Bochenek
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Collin McGladrigan
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Amy Nguyen
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Samuel Norcross
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Yuhan Zhu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Crystal Shuo Shan
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Reed Hausser
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Kevin M Shakesheff
- Division of Regenerative Medicine and Cellular Therapies, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, 02138, USA
- Harvard-MIT Division of Health Science Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, 02138, USA
- Harvard-MIT Division of Health Science Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| |
Collapse
|
26
|
Addison P, Fatakhova K, Rodriguez Rilo HL. Considerations for an Alternative Site of Islet Cell Transplantation. J Diabetes Sci Technol 2020; 14:338-344. [PMID: 31394934 PMCID: PMC7196852 DOI: 10.1177/1932296819868495] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Islet cell transplantation has been limited most by poor graft survival. Optimizing the site of transplantation could improve clinical outcomes by minimizing required donor cells, increasing graft integration, and simplifying the transplantation and monitoring process. In this article, we review the history and significant human and animal data for clinically relevant sites, including the liver, spleen, and kidney subcapsule, and identify promising new sites for further research. While the liver was the first studied site and has been used the most in clinical practice, the majority of transplanted islets become necrotic. We review the potential causes for graft death, including the instant blood-mediated inflammatory reaction, exposure to immunosuppressive agents, and low oxygen tension. Significant research exists on alternative sites for islet cell transplantation, suggesting a promising future for patients undergoing pancreatectomy.
Collapse
Affiliation(s)
- Poppy Addison
- Donald and Barbara Zucker School of
Medicine at Hofstra/Northwell, Hempstead, NY, USA
- Pancreas Disease Center, Northwell
Health System, Manhasset, NY, USA
| | - Karina Fatakhova
- Donald and Barbara Zucker School of
Medicine at Hofstra/Northwell, Hempstead, NY, USA
- Pancreas Disease Center, Northwell
Health System, Manhasset, NY, USA
| | - Horacio L. Rodriguez Rilo
- Donald and Barbara Zucker School of
Medicine at Hofstra/Northwell, Hempstead, NY, USA
- Pancreas Disease Center, Northwell
Health System, Manhasset, NY, USA
- Horacio L. Rodriguez Rilo, MD, Pancreas
Disease Center, 350 Lakeville Road, New Hyde Park, NY 11042, USA.
| |
Collapse
|
27
|
Citro A, Pellegrini S, Dugnani E, Eulberg D, Klussmann S, Piemonti L. CCL2/MCP-1 and CXCL12/SDF-1 blockade by L-aptamers improve pancreatic islet engraftment and survival in mouse. Am J Transplant 2019; 19:3131-3138. [PMID: 31267721 DOI: 10.1111/ajt.15518] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/06/2019] [Accepted: 06/24/2019] [Indexed: 01/25/2023]
Abstract
The blockade of pro-inflammatory mediators is a successful approach to improve the engraftment after islet transplantation. L-aptamers are chemically synthesized, nonimmunogenic bio-stable oligonucleotides that bind and inhibit target molecules conceptually similar to antibodies. We aimed to evaluate if blockade-aptamer-based inhibitors of C-C Motif Chemokine Ligand 2/monocyte chemoattractant protein-1 (CCL2/MCP-1) and C-X-C Motif Chemokine Ligand 12/stromal cell-derived factor-1 (CXCL12/SDF-1) are able to favor islet survival in mouse models for islet transplantation and for type 1 diabetes. We evaluated the efficacy of the CCL2-specific mNOX-E36 and the CXCL12-specific NOX-A12 on islet survival in a syngeneic mouse model of intraportal islet transplantation and in a multiple low doses of streptozotocin (MLD-STZ) diabetes induction model. Moreover, we characterized intrahepatic infiltrated leukocytes by flow cytometry before and 3 days after islet infusion in presence or absence of these inhibitors. The administration for 14 days of mNOX-E36 and NOX-A12 significantly improved islet engraftment, either compound alone or in combination. Intrahepatic islet transplantation recruited CD45+ leucocytes and more specifically CD45+/CD11b+ mono/macrophages; mNOX-E36 and NOX-A12 treatments significantly decreased the recruitment of inflammatory monocytes, CD11b+ /Ly6Chigh /CCR2+ and CD11b+ /Ly6Chigh /CXCR4+ cells, respectively. Additionally, both L-aptamers significantly attenuated diabetes progression in the MLD-STZ model. In conclusion, CCL2/MCP-1 and CXCL12/SDF-1 blockade by L-aptamers is an efficient strategy to improve islet engraftment and survival.
Collapse
Affiliation(s)
- Antonio Citro
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Silvia Pellegrini
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Erica Dugnani
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Sven Klussmann
- NOXXON Pharma AG, Berlin, Germany.,Aptarion Biotech AG, Berlin, Germany
| | - Lorenzo Piemonti
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
28
|
Lankatillake C, Huynh T, Dias DA. Understanding glycaemic control and current approaches for screening antidiabetic natural products from evidence-based medicinal plants. PLANT METHODS 2019; 15:105. [PMID: 31516543 PMCID: PMC6731622 DOI: 10.1186/s13007-019-0487-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/20/2019] [Indexed: 05/15/2023]
Abstract
Type 2 Diabetes Mellitus has reached epidemic proportions as a result of over-nutrition and increasingly sedentary lifestyles. Current therapies, although effective, are not without limitations. These limitations, the alarming increase in the prevalence of diabetes, and the soaring cost of managing diabetes and its complications underscores an urgent need for safer, more efficient and affordable alternative treatments. Over 1200 plant species are reported in ethnomedicine for treating diabetes and these represents an important and promising source for the identification of novel antidiabetic compounds. Evaluating medicinal plants for desirable bioactivity goes hand-in-hand with methods in analytical biochemistry for separating and identifying lead compounds. This review aims to provide a comprehensive summary of current methods used in antidiabetic plant research to form a useful resource for researchers beginning in the field. The review summarises the current understanding of blood glucose regulation and the general mechanisms of action of current antidiabetic medications, and combines knowledge on common experimental approaches for screening plant extracts for antidiabetic activity and currently available analytical methods and technologies for the separation and identification of bioactive natural products. Common in vivo animal models, in vitro models, in silico methods and biochemical assays used for testing the antidiabetic effects of plants are discussed with a particular emphasis on in vitro methods such as cell-based bioassays for screening insulin secretagogues and insulinomimetics. Enzyme inhibition assays and molecular docking are also highlighted. The role of metabolomics, metabolite profiling, and dereplication of data for the high-throughput discovery of novel antidiabetic agents is reviewed. Finally, this review also summarises sample preparation techniques such as liquid-liquid extraction, solid phase extraction, and supercritical fluid extraction, and the critical function of nuclear magnetic resonance and high resolution liquid chromatography-mass spectrometry for the dereplication, putative identification and structure elucidation of natural compounds from evidence-based medicinal plants.
Collapse
Affiliation(s)
- Chintha Lankatillake
- School of Health and Biomedical Sciences, Discipline of Laboratory Medicine, RMIT University, Bundoora, 3083 Australia
| | - Tien Huynh
- School of Science, RMIT University, Bundoora, VIC 3083 Australia
| | - Daniel A. Dias
- School of Health and Biomedical Sciences, Discipline of Laboratory Medicine, RMIT University, Bundoora, 3083 Australia
| |
Collapse
|
29
|
Montanari E, Gonelle-Gispert C, Seebach JD, Knoll MF, Bottino R, Bühler LH. Immunological aspects of allogeneic pancreatic islet transplantation: a comparison between mouse and human. Transpl Int 2019; 32:903-912. [PMID: 31033036 DOI: 10.1111/tri.13445] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 10/29/2018] [Accepted: 04/23/2019] [Indexed: 11/30/2022]
Abstract
Pancreatic islet allotransplantation is a treatment for patients with severe forms of type 1 diabetes. As long-term graft function and survival are not yet optimal, additional studies are warranted in order to continue improving transplant outcomes. The mechanisms of islet graft loss and tolerance induction are often studied in murine diabetes models. Despite numerous islet transplantation studies successfully performed over recent years, translation from experimental mouse models to human clinical application remains elusive. This review aims at critically discussing the strengths and limitations of current mouse models of diabetes and experimental islet transplantation. In particular, we will analyze the causes leading to diabetes and compare the immunological mechanisms responsible for rejection between mouse and human. A better understanding of the experimental mouse models should facilitate translation to human clinical application.
Collapse
Affiliation(s)
- Elisa Montanari
- Department of Surgery, Geneva University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Carmen Gonelle-Gispert
- Department of Surgery, Geneva University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Jörg D Seebach
- Division of Immunology and Allergy, Geneva University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Michael F Knoll
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA, USA
| | - Rita Bottino
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA, USA
| | - Leo H Bühler
- Department of Surgery, Geneva University Hospitals and Medical Faculty, Geneva, Switzerland
| |
Collapse
|
30
|
Anti-Inflammatory Strategies in Intrahepatic Islet Transplantation: A Comparative Study in Preclinical Models. Transplantation 2018; 102:240-248. [PMID: 28902069 DOI: 10.1097/tp.0000000000001925] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND The identification of pathway(s) playing a pivotal role in peritransplant detrimental inflammatory events represents the crucial step toward a better management and outcome of pancreatic islet transplanted patients. Recently, we selected the CXCR1/2 inhibition as a relevant strategy in enhancing pancreatic islet survival after transplantation. METHODS Here, the most clinically used anti-inflammatory compounds (IL1-receptor antagonist, steroids, and TNF-α inhibitor) alone or in combination with a CXCR1/2 inhibitor were evaluated in their ability to improve engraftment or delay graft rejection. To rule out bias related to transplantation site, we used well-established preclinical syngeneic (250 C57BL/6 equivalent islets in C57BL/6) and allogeneic (400 Balb/c equivalent islets in C57BL6) intrahepatic islet transplantation platforms. RESULTS In mice, we confirmed that targeting the CXCR1/2 pathway is crucial in preserving islet function and improving engraftment. In the allogeneic setting, CXCR1/2 inhibitor alone could reduce the overall recruitment of transplant-induced leukocytes and significantly prolong the time to graft rejection both as a single agent and in combination with immunosuppression. No other anti-inflammatory compounds tested (IL1-receptor antagonist, steroids, and TNF-α inhibitor) alone or in combination with CXCR1/2 inhibitor improve islet engraftment and significantly delay graft rejection in the presence of MMF + FK-506 immunosuppressive treatment. CONCLUSIONS These findings indicate that only the CXCR1/2-mediated axis plays a crucial role in controlling the islet damage and should be a target for intervention to improve the efficiency of islet transplantation.
Collapse
|
31
|
Jofra T, Galvani G, Georgia F, Silvia G, Gagliani N, Battaglia M. Murine Pancreatic Islets Transplantation under the Kidney Capsule. Bio Protoc 2018; 8:e2743. [PMID: 34179271 DOI: 10.21769/bioprotoc.2743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/06/2018] [Accepted: 02/06/2018] [Indexed: 11/02/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease caused by the lack of insulin-producing pancreatic beta cells leading to systemic hyperglycemia. Pancreatic islet transplantation is a valid therapeutic approach to restore insulin loss and to promote adequate glycemic control. Pancreatic islet transplantation in mice is an optimal preclinical model to identify new therapeutic strategies aiming at preventing rejection and optimizing post-transplant immuno-suppressive/-tolerogenic therapies. Islet transplantation in preclinical animal models can be performed in different sites such the kidney capsule, spleen, bone marrow and pancreas. This protocol describes murine islet transplantation under the kidney capsule. This is a widely accepted procedure for research purposes. Stress caused in the animals is minimal and it leads to reliable and reproducible results.
Collapse
Affiliation(s)
- Tatiana Jofra
- Diabetes Research Institute (DRI), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giuseppe Galvani
- Diabetes Research Institute (DRI), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fousteri Georgia
- Diabetes Research Institute (DRI), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gregori Silvia
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicola Gagliani
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Medical Department, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuela Battaglia
- Diabetes Research Institute (DRI), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
32
|
Transplant Site Influences the Immune Response After Islet Transplantation: Bone Marrow Versus Liver. Transplantation 2017; 101:1046-1055. [PMID: 27575689 DOI: 10.1097/tp.0000000000001462] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND The aim of this study was to characterize the immune response against intrabone marrow (BM-Tx) or intraliver (liver-Tx) transplanted islets in the presence or in the absence of immunosuppression. METHODS Less (C57BL/6 in Balb/c) and highly (Balb/c in C57BL/6) stringent major histocompatibility complex fully mismatched mouse models were used to evaluate the alloimmune response. Single antigen-mismatched mouse model (C57BL/6 RIP-GP in C57BL/6) was used to evaluate the antigen-specific immune response. Mice received tacrolimus (FK-506, 0.1 mg/kg per day)/mycophenolate mofetil (MMF, 60 mg/kg per day), and anti-CD3 (50 μg/day) either alone or in combination. RESULTS Transplant site did not impact the timing nor the kinetics of the alloimmune and single antigen-specific memory T cell responses in the absence of immunosuppression or in the presence of MMF/FK-506 combination. On the other hand, the median time to graft rejection was 28 ± 5.2 days and 16 ± 2.6 days (P = 0.14) in the presence of anti-CD3 treatment, 50 ± 12.5 days and 10 ± 1.3 days (P = 0.003) in the presence of anti-CD3/MMF/FK-506 treatment for liver-Tx and BM-Tx, respectively. Anti-CD3 did not differentially reach BM and liver tissues but was more effective in reducing graft associated T cell responses in liver-Tx than in BM-Tx. CONCLUSIONS Islets infused in the BM appear less protected from the adaptive immune response in the presence of the anti-CD3 treatment. This result raises some concerns over the potential of the BM as a site for islet allotransplantation.
Collapse
|
33
|
Bishop NH, Nelsen MK, Beard KS, Coulombe M, Gill RG. Differential Impact of Chronic Hyperglycemia on Humoral Versus Cellular Primary Alloimmunity. Diabetes 2017; 66:981-986. [PMID: 28069641 PMCID: PMC5360296 DOI: 10.2337/db16-0218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 01/04/2017] [Indexed: 02/07/2023]
Abstract
Diabetes is prevalent among solid organ transplant recipients and is universal among islet transplant recipients. Whereas diabetes is often considered to result in an immune-compromised state, the impact of chronic hyperglycemia on host alloimmunity is not clear. Potential immune-modifying effects of obesity, autoimmunity, or diabetogenic agents like streptozotocin may confound understanding alloimmunity in experimental models of diabetes. Therefore, we sought to determine the role of chronic hyperglycemia due to insulinopenia on alloimmunity using the nonautoimmune, spontaneously diabetic H-2b-expressing C57BL/6 Ins2Akita mice (Akita). Akita mice harbor a mutated Ins2 allele that dominantly suppresses insulin secretion, resulting in lifelong diabetes. We used BALB/c donors (H-2d) to assess alloimmunization and islet transplantation outcomes in Akita recipients. Surprisingly, chronic hyperglycemia had little effect on primary T-cell reactivity after alloimmunization. Moreover, Akita mice readily rejected islet allografts, and chronic hyperglycemia had no impact on the magnitude or quality of intragraft T-cell responses. In contrast, allospecific IgM and IgG were significantly decreased in Akita mice after alloimmunization. Thus, whereas diabetes influences host immune defense, hyperglycemia itself does not cause generalized alloimmune impairment. Our data suggest that immune compromise in diabetes due to hyperglycemia may not apply to cellular rejection of transplants.
Collapse
Affiliation(s)
- Nicholas H Bishop
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Michelle K Nelsen
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - K Scott Beard
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Marilyne Coulombe
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Ronald G Gill
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
34
|
Bender C, Christen S, Scholich K, Bayer M, Pfeilschifter JM, Hintermann E, Christen U. Islet-Expressed CXCL10 Promotes Autoimmune Destruction of Islet Isografts in Mice With Type 1 Diabetes. Diabetes 2017; 66:113-126. [PMID: 27797910 DOI: 10.2337/db16-0547] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 10/21/2016] [Indexed: 11/13/2022]
Abstract
Type 1 diabetes (T1D) results from the autoimmune destruction of insulin-producing β-cells in the pancreas. Thereby, the chemokine CXC-motif ligand 10 (CXCL10) plays an important role in the recruitment of autoaggressive lymphocytes to the islets of Langerhans. Transplantation of isolated islets as a promising therapy for T1D has been hampered by early graft rejection. Here, we investigated the influence of CXCL10 on the autoimmune destruction of islet isografts using RIP-LCMV mice expressing a lymphocytic choriomeningitis virus (LCMV) protein in the β-cells. RIP-LCMV islets express CXCL10 after isolation and maintain CXCL10 production after engraftment. Thus, we isolated islets from either normal or CXCL10-deficient RIP-LCMV mice and transferred them under the kidney capsule of diabetic RIP-LCMV mice. We found that the autoimmune destruction of CXCL10-deficient islet isografts was significantly reduced. The autoimmune destruction was also diminished in mice administered with an anti-CXCL10 antibody. The persistent protection from autoimmune destruction was paralleled by an increase in FoxP3+ regulatory T cells within the cellular infiltrates around the islet isografts. Consequently, CXCL10 might influence the cellular composition locally in the islet graft, thereby playing a role in the autoimmune destruction. CXCL10 might therefore constitute a potential therapeutic target to prolong islet graft survival.
Collapse
Affiliation(s)
- Christine Bender
- Institute for Pharmacology and Toxicology, Pharmazentrum Frankfurt/Center for Drug Research, Development, and Safety (ZAFES), Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Selina Christen
- Institute for Pharmacology and Toxicology, Pharmazentrum Frankfurt/Center for Drug Research, Development, and Safety (ZAFES), Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Klaus Scholich
- Institute for Clinical Pharmacology, Pharmazentrum Frankfurt/Center for Drug Research, Development, and Safety (ZAFES), Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Monika Bayer
- Institute for Pharmacology and Toxicology, Pharmazentrum Frankfurt/Center for Drug Research, Development, and Safety (ZAFES), Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Josef M Pfeilschifter
- Institute for Pharmacology and Toxicology, Pharmazentrum Frankfurt/Center for Drug Research, Development, and Safety (ZAFES), Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Edith Hintermann
- Institute for Pharmacology and Toxicology, Pharmazentrum Frankfurt/Center for Drug Research, Development, and Safety (ZAFES), Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Urs Christen
- Institute for Pharmacology and Toxicology, Pharmazentrum Frankfurt/Center for Drug Research, Development, and Safety (ZAFES), Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
35
|
King A, Bowe J. Animal models for diabetes: Understanding the pathogenesis and finding new treatments. Biochem Pharmacol 2015; 99:1-10. [PMID: 26432954 DOI: 10.1016/j.bcp.2015.08.108] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 08/26/2015] [Indexed: 02/06/2023]
Abstract
Diabetes mellitus is a lifelong, metabolic disease that is characterised by an inability to maintain normal glucose homeostasis. There are several different forms of diabetes, however the two most common are Type 1 and Type 2 diabetes. Type 1 diabetes is caused by the autoimmune destruction of pancreatic beta cells and a subsequent lack of insulin production, whilst Type 2 diabetes is due to a combination of both insulin resistance and an inability of the beta cells to compensate adequately with increased insulin release. Animal models are increasingly being used to elucidate the mechanisms underlying both Type 1 and Type 2 diabetes as well as to identify and refine novel treatments. However, a wide range of different animal models are currently in use. The majority of these models are suited to addressing certain specific aspects of diabetes research, but may be of little use in other studies. All have pros and cons, and selecting an appropriate model for addressing a specific question is not always a trivial task and will influence the study results and their interpretation. Thus, as the number of available animal models increases it is important to consider the potential roles of these models in the many different aspects of diabetes research. This review gathers information on the currently used experimental animal models of both Type 1 and Type 2 diabetes and evaluates their advantages and disadvantages for research purposes and details the factors that should be taken into account in their use.
Collapse
Affiliation(s)
- Aileen King
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Hodgkin Building 2nd Floor, Guy's Campus, King's College London, London SE1 1UL, United Kingdom.
| | - James Bowe
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Hodgkin Building 2nd Floor, Guy's Campus, King's College London, London SE1 1UL, United Kingdom
| |
Collapse
|
36
|
King K, Rosenthal A. The adverse effects of diabetes on osteoarthritis: update on clinical evidence and molecular mechanisms. Osteoarthritis Cartilage 2015; 23:841-50. [PMID: 25837996 PMCID: PMC5530368 DOI: 10.1016/j.joca.2015.03.031] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 03/02/2015] [Accepted: 03/16/2015] [Indexed: 02/02/2023]
Abstract
Projected increases in the prevalence of both diabetes mellitus (DM) and osteoarthritis (OA) ensure their common co-existence. In an era of increasing attention to personalized medicine, understanding the influence of common comorbidities such as DM should result in improved care of patients with OA. In this narrative review, we summarize the literature addressing the interactions between DM and OA spanning the years from 1962 to 2014. We separated studies depending on whether they investigated clinical populations, animal models, or cells and tissues. The clinical literature addressing the influence of DM on OA and its therapeutic outcomes suggests that DM may augment the development and severity of OA and that DM increases risks associated with joint replacement surgery. The few high quality studies using animal models also support an adverse effect of DM on OA. We review strengths and weaknesses of the common rodent models of DM. The heterogeneous literature derived from studies of articular cells and tissues also supports the existence of biochemical and biomechanical changes in articular tissues in DM, and begins to characterize molecular mechanisms activated in diabetic-like environs which may contribute to OA. Increasing evidence from the clinic and the laboratory supports an adverse effect of DM on the development, severity, and therapeutic outcomes for OA. To understand the mechanisms through which DM contributes to OA, further studies are clearly necessary. Future studies of DM-influenced mechanisms may shed light on general mechanisms of OA pathogenesis and result in more specific and effective therapies for all OA patients.
Collapse
Affiliation(s)
- K.B. King
- Department of Orthopaedics, University of Colorado School of Medicine, Aurora, CO, USA,Surgical Service, Orthopaedic Service, Eastern Colorado Health Care System, Veterans Affairs, Denver, CO, USA
| | - A.K. Rosenthal
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA,Medicine Service, Rheumatology Service, The Clement J. Zablocki Medical Center, Veterans Affairs, Milwaukee, WI, USA,Address correspondence and reprint requests to: A.K. Rosenthal, Zablocki VA Medical Center, 5000 W. National Avenue, Milwaukee, WI 53295-1000, USA. Tel: 1-(414)-955-7027; Fax: 1-(414)-955-6205
| |
Collapse
|
37
|
Engraftment versus immunosuppression: cost-benefit analysis of immunosuppression after intrahepatic murine islet transplantation. Transplantation 2014; 97:1019-26. [PMID: 24770621 DOI: 10.1097/tp.0000000000000104] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Immunosuppression (IS) in islet transplantation (Tx) is a double-edged sword: it prevents immunoreaction but has the potential to impair islet engraftment. The aim of this study was to identify in murine animal models the IS platform with the best balance between these two opposite effects. METHODS To study the impact of IS on islet engraftment diabetic C57BL/6 mice were transplanted with 350 syngeneic islets through the portal vein and treated once-daily with either rapamycin (RAPA; 0.1-0.5-1 mg/kg ip), tacrolimus (FK506; 0.1-0.5-1 mg/kg ip), mycophenolate mofetil (MMF; 60-120-300 mg/kg oral) or vehicle for 14 days. Islet function was evaluated by measuring not-fasting glycemia and by performing an IVGTT on days 15 and 30 post-Tx. RESULTS RAPA ≥0.5 mg/Kg, FK506 ≥0.5 mg/Kg, and MMF ≥120 mg/kg had detrimental effects on islet engraftment but not on the function of islets already engrafted in the liver. The effect on engraftment was irreversible and persisted even after IS withdrawal. The lower dose of IS that did not affect engraftment was tested for preventing rejection in the full mismatch allogeneic Tx BALB/c to C57BL/6 model. RAPA and/or FK506 were inefficient in preventing rejection, even when anti-IL2R mAb was added to the IS regimen. On the other hand, MMF alone or in association with FK506 significantly prolonged the time to islet rejection. CONCLUSION IS showed profound dose-dependent deleterious effects on islet cell engraftment. The MMF/FK506 combination proved the best balance with less toxicity at the time of engraftment and more efficacy in controlling graft rejection.
Collapse
|