1
|
Chuang ST, Alcazar O, Watts B, Abdulreda MH, Buchwald P. Small-molecule inhibitors of the CD40-CD40L costimulatory interaction are effective in pancreatic islet transplantation and prevention of type 1 diabetes models. Front Immunol 2024; 15:1484425. [PMID: 39606229 PMCID: PMC11599200 DOI: 10.3389/fimmu.2024.1484425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
As part of our work to develop small-molecule inhibitors (SMIs) of the CD40-CD40L(CD154) costimulatory protein-protein interaction, here, we describe the ability of two of our most promising SMIs, DRI-C21041 and DRI-C21095, to prolong the survival and function of islet allografts in two murine models of islet transplantation (under the kidney capsule and in the anterior chamber of the eye) and to prevent autoimmune type 1 diabetes (T1D) onset in NOD mice. In both transplant models, a significant portion of islet allografts (50%-80%) remained intact and functional long after terminating treatment, suggesting the possibility of inducing operational immune tolerance via inhibition of the CD40-CD40L axis. SMI-treated mice maintained the structural integrity and function of their islet allografts with concomitant reduction in immune cell infiltration as evidenced by direct longitudinal imaging in situ. Furthermore, in female NODs, three-month SMI treatment reduced the incidence of diabetes from 80% to 60% (DRI-C21041) and 25% (DRI-C21095). These results (i) demonstrate the susceptibility of this TNF superfamily protein-protein interaction to small-molecule inhibition, (ii) confirm the in vivo therapeutic potential of these SMIs of a critical immune checkpoint, and (iii) reaffirm the therapeutic promise of CD40-CD40L blockade in islet transplantation and T1D prevention. Thus, CD40L-targeting SMIs could ultimately lead to alternative immunomodulatory therapeutics for transplant recipients and prevention of autoimmune diseases that are safer, less immunogenic, more controllable (shorter half-lives), and more patient-friendly (i.e., suitable for oral administration, which makes them easier to administer) than corresponding antibody-based interventions.
Collapse
Affiliation(s)
- Sung-Ting Chuang
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Oscar Alcazar
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Brandon Watts
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Midhat H. Abdulreda
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Ophthalmology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
2
|
Yang SN, Shi Y, Berggren PO. The anterior chamber of the eye technology and its anatomical, optical, and immunological bases. Physiol Rev 2024; 104:881-929. [PMID: 38206586 PMCID: PMC11381035 DOI: 10.1152/physrev.00024.2023] [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: 06/20/2023] [Revised: 11/30/2023] [Accepted: 01/05/2024] [Indexed: 01/12/2024] Open
Abstract
The anterior chamber of the eye (ACE) is distinct in its anatomy, optics, and immunology. This guarantees that the eye perceives visual information in the context of physiology even when encountering adverse incidents like inflammation. In addition, this endows the ACE with the special nursery bed iris enriched in vasculatures and nerves. The ACE constitutes a confined space enclosing an oxygen/nutrient-rich, immune-privileged, and less stressful milieu as well as an optically transparent medium. Therefore, aside from visual perception, the ACE unexpectedly serves as an excellent transplantation site for different body parts and a unique platform for noninvasive, longitudinal, and intravital microimaging of different grafts. On the basis of these merits, the ACE technology has evolved from the prototypical through the conventional to the advanced version. Studies using this technology as a versatile biomedical research platform have led to a diverse range of basic knowledge and in-depth understanding of a variety of cells, tissues, and organs as well as artificial biomaterials, pharmaceuticals, and abiotic substances. Remarkably, the technology turns in vivo dynamic imaging of the morphological characteristics, organotypic features, developmental fates, and specific functions of intracameral grafts into reality under physiological and pathological conditions. Here we review the anatomical, optical, and immunological bases as well as technical details of the ACE technology. Moreover, we discuss major achievements obtained and potential prospective avenues for this technology.
Collapse
Affiliation(s)
- Shao-Nian Yang
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Yue Shi
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
3
|
Wardell CM, Fung VC, Chen E, Haque M, Gillies J, Spanier JA, Mojibian M, Fife BT, Levings MK. Short Report: CAR Tregs mediate linked suppression and infectious tolerance in islet transplantation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.06.588414. [PMID: 38645184 PMCID: PMC11030375 DOI: 10.1101/2024.04.06.588414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Regulatory T cells (Tregs) have potential as a cell-based therapy to prevent or treat transplant rejection and autoimmunity. Using an HLA-A2-specific chimeric antigen receptor (A2-CAR), we previously showed that adoptive transfer of A2-CAR Tregs limited anti-HLA-A2 alloimmunity. However, it was unknown if A2-CAR Tregs could also limit immunity to autoantigens. Using a model of HLA-A2+ islet transplantation into immunodeficient non-obese diabetic mice, we investigated if A2-CAR Tregs could control diabetes induced by islet-autoreactive (BDC2.5) T cells. In mice transplanted with HLA-A2+ islets, A2-CAR Tregs reduced BDC2.5 T cell engraftment, proliferation and cytokine production, and protected mice from diabetes. Tolerance to islets was systemic, including protection of the HLA-A2negative endogenous pancreas. In tolerant mice, a significant proportion of BDC2.5 T cells gained FOXP3 expression suggesting that long-term tolerance is maintained by de novo Treg generation. Thus, A2-CAR Tregs mediate linked suppression and infectious tolerance and have potential therapeutic use to simultaneously control both allo- and autoimmunity in islet transplantation.
Collapse
Affiliation(s)
- Christine M. Wardell
- BC Children’s Hospital Research Institute, University of British Columbia; Vancouver, BC, Canada
- Dept of Surgery, University of British Columbia; Vancouver, BC, Canada
| | - Vivian C.W. Fung
- BC Children’s Hospital Research Institute, University of British Columbia; Vancouver, BC, Canada
- Dept of Surgery, University of British Columbia; Vancouver, BC, Canada
| | - Eleanor Chen
- BC Children’s Hospital Research Institute, University of British Columbia; Vancouver, BC, Canada
- Dept of Surgery, University of British Columbia; Vancouver, BC, Canada
| | - Manjurul Haque
- BC Children’s Hospital Research Institute, University of British Columbia; Vancouver, BC, Canada
- Dept of Surgery, University of British Columbia; Vancouver, BC, Canada
| | - Jana Gillies
- BC Children’s Hospital Research Institute, University of British Columbia; Vancouver, BC, Canada
- Dept of Surgery, University of British Columbia; Vancouver, BC, Canada
| | - Justin A. Spanier
- Center for Immunology, University of Minnesota Medical School; Minneapolis, MN, USA
- Center for Autoimmune Disease Research, Department of Medicine, University of Minnesota Medical School; Minneapolis, MN, USA
- Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School; Minneapolis, MN, USA
| | - Majid Mojibian
- BC Children’s Hospital Research Institute, University of British Columbia; Vancouver, BC, Canada
- Dept of Surgery, University of British Columbia; Vancouver, BC, Canada
| | - Brian T. Fife
- Center for Immunology, University of Minnesota Medical School; Minneapolis, MN, USA
- Center for Autoimmune Disease Research, Department of Medicine, University of Minnesota Medical School; Minneapolis, MN, USA
- Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School; Minneapolis, MN, USA
| | - Megan K. Levings
- BC Children’s Hospital Research Institute, University of British Columbia; Vancouver, BC, Canada
- Dept of Surgery, University of British Columbia; Vancouver, BC, Canada
- School of Biomedical Engineering, University of British Columbia; Vancouver, BC, Canada
| |
Collapse
|
4
|
Li S, Yuan H, Yang K, Li Q, Xiang M. Pancreatic sympathetic innervation disturbance in type 1 diabetes. Clin Immunol 2023; 250:109319. [PMID: 37024024 DOI: 10.1016/j.clim.2023.109319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/15/2023] [Accepted: 03/06/2023] [Indexed: 04/08/2023]
Abstract
Pancreatic sympathetic innervation can directly affect the function of islet. The disorder of sympathetic innervation in islets during the occurrence of type 1 diabetes (T1D) has been reported to be controversial with the inducing factor unclarified. Several studies have uncovered the critical role that sympathetic signals play in controlling the local immune system. The survival and operation of endocrine cells can be regulated by immune cell infiltration in islets. In the current review, we focused on the impact of sympathetic signals working on islets cell regulation, and discussed the potential factors that can induce the sympathetic innervation disorder in the islets. We also summarized the effect of interference with the islet sympathetic signals on the T1D occurrence. Overall, complete understanding of the regulatory effect of sympathetic signals on islet cells and local immune system could facilitate to design better strategies to control inflammation and protect β cells in T1D therapy.
Collapse
Affiliation(s)
- Senlin Li
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huimin Yuan
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Keshan Yang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qing Li
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ming Xiang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
5
|
Fluorescence Angiography with Dual Fluorescence for the Early Detection and Longitudinal Quantitation of Vascular Leakage in Retinopathy. Biomedicines 2023; 11:biomedicines11020293. [PMID: 36830829 PMCID: PMC9953145 DOI: 10.3390/biomedicines11020293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/03/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Diabetic retinopathy (DR) afflicts more than 93 million people worldwide and is a leading cause of vision loss in working adults. While DR therapies are available, early DR development may go undetected without treatment due to the lack of sufficiently sensitive tools. Therefore, early detection is critically important to enable efficient treatment before progression to vision-threatening complications. A major clinical manifestation of early DR is retinal vascular leakage that may progress from diffuse to more localized focal leakage, leading to increased retinal thickness and diabetic macular edema (DME). In preclinical research, a hallmark of DR in mouse models is diffuse retinal leakage without increased thickness or DME, which limits the utility of optical coherence tomography and fluorescein angiography (FA) for early detection. The Evans blue assay detects diffuse leakage but requires euthanasia, which precludes longitudinal studies in the same animals. METHODS We developed a new modality of ratiometric fluorescence angiography with dual fluorescence (FA-DF) to reliably detect and longitudinally quantify diffuse retinal vascular leakage in mouse models of induced and spontaneous DR. RESULTS These studies demonstrated the feasibility and sensitivity of FA-DF in detecting and quantifying retinal vascular leakage in the same mice over time during DR progression in association with chronic hyperglycemia and age. CONCLUSIONS These proof-of-concept studies demonstrated the promise of FA-DF as a minimally invasive method to quantify DR leakage in preclinical mouse models longitudinally.
Collapse
|
6
|
Park WY, Kim J, Le H, Kim B, Berggren PO, Kim KH. Longitudinal monitoring of pancreatic islet damage in streptozotocin-treated mice with optical coherence microscopy. BIOMEDICAL OPTICS EXPRESS 2023; 14:54-64. [PMID: 36698658 PMCID: PMC9841987 DOI: 10.1364/boe.470188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/24/2022] [Accepted: 11/27/2022] [Indexed: 06/17/2023]
Abstract
Pancreatic islets regulate glucose homeostasis in the body, and their dysfunction is closely related to diabetes. Islet transplantation into the anterior chamber of the eye (ACE) was recently developed for both in vivo islet study and diabetes treatment. Optical coherence microscopy (OCM) was previously used to monitor ACE transplanted islets in non-obese diabetic (NOD) mice for detecting autoimmune attack. In this study, OCM was applied to streptozotocin (STZ)-induced diabetic mouse models for the early detection of islet damage. A custom extended-focus OCM (xfOCM) was used to image islet grafts in the ACE longitudinally during STZ-induced beta cell destruction together with conventional bright-field (BF) imaging and invasive glucose level measurement. xfOCM detected local structural changes and vascular degradation during the islet damage which was confirmed by confocal imaging of extracted islet grafts. xfOCM detection of islet damage was more sensitive than BF imaging and glucose measurement. Longitudinal xfOCM images of islet grafts were quantitatively analyzed. All these results showed that xfOCM could be used as a non-invasive and sensitive monitoring method for the early detection of deficient islet grafts in the ACE with potential applications to human subjects.
Collapse
Affiliation(s)
- Won Yeong Park
- Department of Mechanical Engineering, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Jaeyoon Kim
- Division of Integrative Biosciences & Biotechnology, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Hoan Le
- Department of Mechanical Engineering, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Bumju Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Per-Olof Berggren
- Division of Integrative Biosciences & Biotechnology, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-17 76 Stockholm, Sweden
| | - Ki Hean Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| |
Collapse
|
7
|
Wagner LE, Melnyk O, Duffett BE, Linnemann AK. Mouse models and human islet transplantation sites for intravital imaging. Front Endocrinol (Lausanne) 2022; 13:992540. [PMID: 36277698 PMCID: PMC9579277 DOI: 10.3389/fendo.2022.992540] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/09/2022] [Indexed: 01/12/2023] Open
Abstract
Human islet transplantations into rodent models are an essential tool to aid in the development and testing of islet and cellular-based therapies for diabetes prevention and treatment. Through the ability to evaluate human islets in an in vivo setting, these studies allow for experimental approaches to answer questions surrounding normal and disease pathophysiology that cannot be answered using other in vitro and in vivo techniques alone. Intravital microscopy enables imaging of tissues in living organisms with dynamic temporal resolution and can be employed to measure biological processes in transplanted human islets revealing how experimental variables can influence engraftment, and transplant survival and function. A key consideration in experimental design for transplant imaging is the surgical placement site, which is guided by the presence of vasculature to aid in functional engraftment of the islets and promote their survival. Here, we review transplantation sites and mouse models used to study beta cell biology in vivo using intravital microscopy and we highlight fundamental observations made possible using this methodology.
Collapse
Affiliation(s)
- Leslie E. Wagner
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Olha Melnyk
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Bryce E. Duffett
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Amelia K. Linnemann
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
8
|
Tun SBB, Chua M, Tan GSW, Leibiger I, Ali Y, Barathi VA, Berggren PO. Local Dexamethasone Administration Delays Allogeneic Islet Graft Rejection in the Anterior Chamber of the Eye of Non-Human Primates. Cell Transplant 2022; 31:9636897221098038. [PMID: 35603580 PMCID: PMC9125106 DOI: 10.1177/09636897221098038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pancreatic islet transplantation into the anterior chamber of the eye (ACE) has been shown to improve glycemic control and metabolic parameters of diabetes in both murine and primate models. This novel transplantation site also allows the delivery of therapeutic agents, such as immunosuppressive drugs, locally to prevent islet graft rejection and circumvent unwanted systemic side effects. Local intravitreal administration of micronized dexamethasone implant was performed prior to allogeneic islet transplantation into the ACEs of non-human primates. Two study groups were observed namely allogeneic graft without immunosuppression (n = 4 eyes) and allogeneic graft with local immunosuppression (n = 8 eyes). Survival of islet grafts and dexamethasone concentration in the ACE were assessed in parallel for 24 weeks. Allogeneic islet grafts with local dexamethasone treatment showed significantly better survival than those with no immunosuppression (median survival time- 15 weeks vs 3 weeks, log-rank test p<0.0001). Around 73% of the grafts still survived at week 10 with a single local dexamethasone implant, where the control group showed no graft survival. Dexamethasone treated islet grafts revealed a good functional response to high glucose stimulation despite there was a transient suppression of insulin secretion from week 8 to 12. Our findings show a significant improvement of allografts survival in the ACE with local dexamethasone treatment. These results highlight the feasibility of local administration of pharmacological compounds in the ACE to improve islet graft survival and function. By eliminating the need for systemic immunosuppression, these findings may impact clinical islet transplantation in the treatment of diabetes, and the ACE may serve as a novel therapeutic islet transplantation site with high potential for local pharmacological intervention.
Collapse
Affiliation(s)
- Sai Bo Bo Tun
- Translational Pre-Clinical Model Platform, Singapore Eye Research Institute, Singapore
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Minni Chua
- Translational Pre-Clinical Model Platform, Singapore Eye Research Institute, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Gavin Siew Wei Tan
- Translational Pre-Clinical Model Platform, Singapore Eye Research Institute, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, DUKE-NUS Medical School, Singapore
| | - Ingo Leibiger
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Yusuf Ali
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Veluchamy Amutha Barathi
- Translational Pre-Clinical Model Platform, Singapore Eye Research Institute, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, DUKE-NUS Medical School, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Per-Olof Berggren
- Translational Pre-Clinical Model Platform, Singapore Eye Research Institute, Singapore
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
9
|
Jacobelli J, Buser AE, Heiden DL, Friedman RS. Autoimmunity in motion: Mechanisms of immune regulation and destruction revealed by in vivo imaging. Immunol Rev 2022; 306:181-199. [PMID: 34825390 PMCID: PMC9135487 DOI: 10.1111/imr.13043] [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: 11/01/2021] [Accepted: 11/06/2021] [Indexed: 11/30/2022]
Abstract
Autoimmunity arises when mechanisms of immune tolerance fail. Here we discuss mechanisms of T cell activation and tolerance and the dynamics of the autoimmune response at the site of disease. Live imaging of autoimmunity provides the ability to analyze immune cell dynamics at the single-cell level within the complex intact environment where disease occurs. These analyses have revealed mechanisms of T cell activation and tolerance in the lymph nodes, mechanisms of T cell entry into sites of autoimmune disease, and mechanisms leading to pathogenesis or protection in the autoimmune lesions. The overarching conclusions point to stable versus transient T cell antigen presenting cell interactions dictating the balance between T cell activation and tolerance, and T cell restimulation as a driver of pathogenesis at the site of autoimmunity. Findings from models of multiple sclerosis and type 1 diabetes are highlighted, however, the results have implications for basic mechanisms of T cell regulation during immune responses, tumor immunity, and autoimmunity.
Collapse
Affiliation(s)
- Jordan Jacobelli
- Barbara Davis Center for Diabetes, Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Alan E. Buser
- Barbara Davis Center for Diabetes, Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Dustin L. Heiden
- Barbara Davis Center for Diabetes, Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Rachel S. Friedman
- Barbara Davis Center for Diabetes, Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
10
|
Zhao K, Shi Y, Yu J, Yu L, Mael A, Li Y, Kolton A, Joyce T, Odorico J, Berggren PO, Yang SN. Intracameral Microimaging of Maturation of Human iPSC Derivatives into Islet Endocrine Cells. Cell Transplant 2022; 31:9636897211066508. [PMID: 35156411 PMCID: PMC8848082 DOI: 10.1177/09636897211066508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
We exploited the anterior chamber of the eye (ACE) of immunodeficient mice as an ectopic site for both transplantation and microimaging of engineered surrogate islets from human induced pluripotent stem cells (hiPSC-SIs). These islets contained a majority of insulin-expressing cells, positive or negative for PDX1 and NKX6.1, and a minority of glucagon- or somatostatin-positive cells. Single, non-aggregated hiPSC-SIs were satisfactorily engrafted onto the iris. They underwent gradual vascularization and progressively increased their light scattering signals, reflecting the abundance of zinc-insulin crystal packaged inside mature insulin secretory granules. Intracameral hiPSC-SIs retrieved from recipients showed enhanced insulin immunofluorescence in correlation with the parallel increase in overall vascularization and light backscattering during the post-transplantation period. This approach enables longitudinal, nondestructive and intravital microimaging of cell fates, engraftment, vascularization and mature insulin secretory granules of single hiPSC-SI grafts, and may offer a feasible and reliable means to screen compounds for promoting in vivo hiPSC-SI maturation.
Collapse
Affiliation(s)
- Kaixuan Zhao
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
| | - Yue Shi
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
| | - Jia Yu
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
| | - Lina Yu
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
| | - Amber Mael
- Regenerative Medical Solutions, Inc., Madison, WI, USA
| | - Yuxin Li
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | | | - Thomas Joyce
- Regenerative Medical Solutions, Inc., Madison, WI, USA
| | - Jon Odorico
- Regenerative Medical Solutions, Inc., Madison, WI, USA
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
| | - Shao-Nian Yang
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| |
Collapse
|
11
|
Abstract
β-Cells in the islet of Langerhans have a central role in maintaining energy homeostasis. Understanding the physiology of β-cells and other islet cells requires a deep understanding of their structural and functional organization, their interaction with vessels and nerves, the layout of paracrine interactions, and the relationship between subcellular compartments and protein complexes inside each cell. These elements are not static; they are dynamic and exert their biological actions at different scales of time. Therefore, scientists must be able to investigate (and visualize) short- and long-lived events within the pancreas and β-cells. Current technological advances in microscopy are able to bridge multiple spatiotemporal scales in biology to reveal the complexity and heterogeneity of β-cell biology. Here, I briefly discuss the historical discoveries that leveraged microscopes to establish the basis of β-cell anatomy and structure, the current imaging platforms that allow the study of islet and β-cell biology at multiple scales of resolution, and their challenges and implications. Lastly, I outline how the remarkable longevity of structural elements at different scales in biology, from molecules to cells to multicellular structures, could represent a previously unrecognized organizational pattern in developing and adult β-cells and pancreas biology.
Collapse
Affiliation(s)
- Rafael Arrojo E Drigo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| |
Collapse
|
12
|
Abdulreda MH, Berggren PO. Challenges in stem cell-derived islet replacement therapy can be overcome. Cell Transplant 2021; 30:9636897211045320. [PMID: 34565192 PMCID: PMC8485158 DOI: 10.1177/09636897211045320] [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] [Indexed: 11/23/2022] Open
Abstract
In this Commentary, we echo the conclusions of a recent review titled
“The promise of stem cell-derived islet replacement
therapy,” which highlighted recent advances in
producing glucose responsive “islets” from stem cells and the benefits
of their use in islet transplant therapy in type 1 diabetes (T1D). The
review also outlined the status of clinical islet transplantation and
the challenges that have prevented it from reaching its full
therapeutic promise. We agree with the conclusions of the review and
suggest that the identified challenges may be overcome by using the
eye anterior chamber as an islet transplant site. We anticipate that
the combination of stem cell-derived islets and intraocular transplant
could help this promising T1D therapy reach full fruition.
Collapse
Affiliation(s)
- Midhat H Abdulreda
- Diabetes Research Institute, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Per-Olof Berggren
- Diabetes Research Institute, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.,The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
| |
Collapse
|
13
|
HGAL inhibits lymphoma dissemination by interacting with multiple Cytoskeletal proteins. Blood Adv 2021; 5:5072-5085. [PMID: 34543391 PMCID: PMC9153012 DOI: 10.1182/bloodadvances.2021004304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 07/09/2021] [Indexed: 12/03/2022] Open
Abstract
In vivo experiments demonstrate that HGAL expression in lymphoma decreases tumor dissemination and prolongs animal survival. HGAL inhibits cell motility by interacting with multiple cytoskeletal proteins, thereby affecting cell motility by multiple mechanisms.
Human germinal center–associated lymphoma (HGAL) is an adaptor protein specifically expressed in germinal center lymphocytes. High expression of HGAL is a predictor of prolonged survival of diffuse large B-cell lymphoma (DLBCL) and classic Hodgkin lymphoma. Furthermore, HGAL expression is associated with early-stage DLBCL, thus potentially limiting lymphoma dissemination. In our previous studies, we demonstrated that HGAL regulates B-cell receptor signaling and cell motility in vitro and deciphered some molecular mechanisms underlying these effects. By using novel animal models for in vivo DLBCL dispersion, we demonstrate here that HGAL decreases lymphoma dissemination and prolongs survival. Furthermore, by using an unbiased proteomic approach, we demonstrate that HGAL may interact with multiple cytoskeletal proteins thereby implicating a multiplicity of effects in regulating lymphoma motility and spread. Specifically, we show that HGAL interacts with tubulin, and this interaction may also contribute to HGAL effects on cell motility. These findings recapitulate previous observations in humans, establish the role of HGAL in dissemination of lymphoma in vivo, and explain improved survival of patients with HGAL-expressing lymphomas.
Collapse
|
14
|
Integrated Metabolomics and Proteomics Analyses in the Local Milieu of Islet Allografts in Rejection versus Tolerance. Int J Mol Sci 2021; 22:ijms22168754. [PMID: 34445459 PMCID: PMC8395897 DOI: 10.3390/ijms22168754] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/03/2021] [Accepted: 08/11/2021] [Indexed: 11/24/2022] Open
Abstract
An understanding of the immune mechanisms that lead to rejection versus tolerance of allogeneic pancreatic islet grafts is of paramount importance, as it facilitates the development of innovative methods to improve the transplant outcome. Here, we used our established intraocular islet transplant model to gain novel insight into changes in the local metabolome and proteome within the islet allograft’s immediate microenvironment in association with immune-mediated rejection or tolerance. We performed integrated metabolomics and proteomics analyses in aqueous humor samples representative of the graft’s microenvironment under each transplant outcome. The results showed that several free amino acids, small primary amines, and soluble proteins related to the Warburg effect were upregulated or downregulated in association with either outcome. In general, the observed shifts in the local metabolite and protein profiles in association with rejection were consistent with established pro-inflammatory metabolic pathways and those observed in association with tolerance were immune regulatory. Taken together, the current findings further support the potential of metabolic reprogramming of immune cells towards immune regulation through targeted pharmacological and dietary interventions against specific metabolic pathways that promote the Warburg effect to prevent the rejection of transplanted islets and promote their immune tolerance.
Collapse
|
15
|
Aldrich VR, Hernandez-Rovira BB, Chandwani A, Abdulreda MH. NOD Mice-Good Model for T1D but Not Without Limitations. Cell Transplant 2021; 29:963689720939127. [PMID: 32762460 PMCID: PMC7563935 DOI: 10.1177/0963689720939127] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The nonobese diabetic (NOD) mouse model of type 1 diabetes (T1D) was discovered by coincidence in the 1980s and has since been widely used in the investigation of T1D and diabetic complications. The current in vivo study was originally designed to prospectively assess whether hyperglycemia onset is associated with physical destruction or functional impairment of beta cells under inflammatory insult during T1D progression in diabetes-prone female NOD mice. Prediabetic 16- to 20-wk-old NOD mice were transplanted with green fluorescent protein (GFP)-expressing reporter islets in the anterior chamber of the eye (ACE) that were monitored longitudinally, in addition to glycemia, with and without immune modulation using anti-CD3 monoclonal antibody therapy. However, there was an early and vigorous immune reaction against the GFP-expressing beta cells that lead to their premature destruction independent of autoimmune T1D development in progressor mice that eventually became hyperglycemic. This immune reaction also occurred in nonprogressor NOD recipients. These findings showed a previously unknown reaction of NOD mice to GFP that prevented achieving the original goals of this study but highlighted a new feature of the NOD mice that should be considered when designing experiments using this model in T1D research.
Collapse
Affiliation(s)
- Virginia R Aldrich
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Barbara B Hernandez-Rovira
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ankit Chandwani
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Midhat H Abdulreda
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
16
|
Ilegems E, Berggren PO. The Eye as a Transplantation Site to Monitor Pancreatic Islet Cell Plasticity. Front Endocrinol (Lausanne) 2021; 12:652853. [PMID: 33967961 PMCID: PMC8104082 DOI: 10.3389/fendo.2021.652853] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/26/2021] [Indexed: 02/05/2023] Open
Abstract
The endocrine cells confined in the islets of Langerhans are responsible for the maintenance of blood glucose homeostasis. In particular, beta cells produce and secrete insulin, an essential hormone regulating glucose uptake and metabolism. An insufficient amount of beta cells or defects in the molecular mechanisms leading to glucose-induced insulin secretion trigger the development of diabetes, a severe disease with epidemic spreading throughout the world. A comprehensive appreciation of the diverse adaptive procedures regulating beta cell mass and function is thus of paramount importance for the understanding of diabetes pathogenesis and for the development of effective therapeutic strategies. While significant findings were obtained by the use of islets isolated from the pancreas, in vitro studies are inherently limited since they lack the many factors influencing pancreatic islet cell function in vivo and do not allow for longitudinal monitoring of islet cell plasticity in the living organism. In this respect a number of imaging methodologies have been developed over the years for the study of islets in situ in the pancreas, a challenging task due to the relatively small size of the islets and their location, scattered throughout the organ. To increase imaging resolution and allow for longitudinal studies in individual islets, another strategy is based on the transplantation of islets into other sites that are more accessible for imaging. In this review we present the anterior chamber of the eye as a transplantation and imaging site for the study of pancreatic islet cell plasticity, and summarize the major research outcomes facilitated by this technological platform.
Collapse
Affiliation(s)
- Erwin Ilegems
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institute, Stockholm, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institute, Stockholm, Sweden
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Center for Diabetes and Metabolism Research, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- School of Biomedical Sciences, Ulster University, Coleraine, United Kingdom
| |
Collapse
|
17
|
Alcazar O, Hernandez LF, Nakayasu ES, Nicora CD, Ansong C, Muehlbauer MJ, Bain JR, Myer CJ, Bhattacharya SK, Buchwald P, Abdulreda MH. Parallel Multi-Omics in High-Risk Subjects for the Identification of Integrated Biomarker Signatures of Type 1 Diabetes. Biomolecules 2021; 11:383. [PMID: 33806609 PMCID: PMC7999903 DOI: 10.3390/biom11030383] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Biomarkers are crucial for detecting early type-1 diabetes (T1D) and preventing significant β-cell loss before the onset of clinical symptoms. Here, we present proof-of-concept studies to demonstrate the potential for identifying integrated biomarker signature(s) of T1D using parallel multi-omics. METHODS Blood from human subjects at high risk for T1D (and healthy controls; n = 4 + 4) was subjected to parallel unlabeled proteomics, metabolomics, lipidomics, and transcriptomics. The integrated dataset was analyzed using Ingenuity Pathway Analysis (IPA) software for disturbances in the at-risk subjects compared to controls. RESULTS The final quadra-omics dataset contained 2292 proteins, 328 miRNAs, 75 metabolites, and 41 lipids that were detected in all samples without exception. Disease/function enrichment analyses consistently indicated increased activation, proliferation, and migration of CD4 T-lymphocytes and macrophages. Integrated molecular network predictions highlighted central involvement and activation of NF-κB, TGF-β, VEGF, arachidonic acid, and arginase, and inhibition of miRNA Let-7a-5p. IPA-predicted candidate biomarkers were used to construct a putative integrated signature containing several miRNAs and metabolite/lipid features in the at-risk subjects. CONCLUSIONS Preliminary parallel quadra-omics provided a comprehensive picture of disturbances in high-risk T1D subjects and highlighted the potential for identifying associated integrated biomarker signatures. With further development and validation in larger cohorts, parallel multi-omics could ultimately facilitate the classification of T1D progressors from non-progressors.
Collapse
Affiliation(s)
- Oscar Alcazar
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (O.A.); (L.F.H.)
| | - Luis F. Hernandez
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (O.A.); (L.F.H.)
| | - Ernesto S. Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (E.S.N.); (C.D.N.); (C.A.)
| | - Carrie D. Nicora
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (E.S.N.); (C.D.N.); (C.A.)
| | - Charles Ansong
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (E.S.N.); (C.D.N.); (C.A.)
| | - Michael J. Muehlbauer
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA; (M.J.M.); (J.R.B.)
| | - James R. Bain
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA; (M.J.M.); (J.R.B.)
| | - Ciara J. Myer
- Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.J.M.); (S.K.B.)
- Miami Integrative Metabolomics Research Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Sanjoy K. Bhattacharya
- Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.J.M.); (S.K.B.)
- Miami Integrative Metabolomics Research Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Peter Buchwald
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (O.A.); (L.F.H.)
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Midhat H. Abdulreda
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (O.A.); (L.F.H.)
- Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.J.M.); (S.K.B.)
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
18
|
Ceballos GA, Hernandez LF, Paredes D, Betancourt LR, Abdulreda MH. A machine learning approach to predict pancreatic islet grafts rejection versus tolerance. PLoS One 2020; 15:e0241925. [PMID: 33152016 PMCID: PMC7644021 DOI: 10.1371/journal.pone.0241925] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 10/22/2020] [Indexed: 02/06/2023] Open
Abstract
The application of artificial intelligence (AI) and machine learning (ML) in biomedical research promises to unlock new information from the vast amounts of data being generated through the delivery of healthcare and the expanding high-throughput research applications. Such information can aid medical diagnoses and reveal various unique patterns of biochemical and immune features that can serve as early disease biomarkers. In this report, we demonstrate the feasibility of using an AI/ML approach in a relatively small dataset to discriminate among three categories of samples obtained from mice that either rejected or tolerated their pancreatic islet allografts following transplant in the anterior chamber of the eye, and from naïve controls. We created a locked software based on a support vector machine (SVM) technique for pattern recognition in electropherograms (EPGs) generated by micellar electrokinetic chromatography and laser induced fluorescence detection (MEKC-LIFD). Predictions were made based only on the aligned EPGs obtained in microliter-size aqueous humor samples representative of the immediate local microenvironment of the islet allografts. The analysis identified discriminative peaks in the EPGs of the three sample categories. Our classifier software was tested with targeted and untargeted peaks. Working with the patterns of untargeted peaks (i.e., based on the whole pattern of EPGs), it was able to achieve a 21 out of 22 positive classification score with a corresponding 95.45% prediction accuracy among the three sample categories, and 100% accuracy between the rejecting and tolerant recipients. These findings demonstrate the feasibility of AI/ML approaches to classify small numbers of samples and they warrant further studies to identify the analytes/biochemicals corresponding to discriminative features as potential biomarkers of islet allograft immune rejection and tolerance.
Collapse
Affiliation(s)
- Gerardo A. Ceballos
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, United States of America
- * E-mail: (GAC); (MHA)
| | - Luis F. Hernandez
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Daniel Paredes
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, United States of America
| | - Luis R. Betancourt
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, United States of America
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Midhat H. Abdulreda
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, United States of America
- * E-mail: (GAC); (MHA)
| |
Collapse
|
19
|
Chitirala P, Chang HF, Martzloff P, Harenberg C, Ravichandran K, Abdulreda MH, Berggren PO, Krause E, Schirra C, Leinders-Zufall T, Benseler F, Brose N, Rettig J. Studying the biology of cytotoxic T lymphocytes in vivo with a fluorescent granzyme B-mTFP knock-in mouse. eLife 2020; 9:e58065. [PMID: 32696761 PMCID: PMC7375811 DOI: 10.7554/elife.58065] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/01/2020] [Indexed: 12/23/2022] Open
Abstract
Understanding T cell function in vivo is of key importance for basic and translational immunology alike. To study T cells in vivo, we developed a new knock-in mouse line, which expresses a fusion protein of granzyme B, a key component of cytotoxic granules involved in T cell-mediated target cell-killing, and monomeric teal fluorescent protein from the endogenous Gzmb locus. Homozygous knock-ins, which are viable and fertile, have cytotoxic T lymphocytes with endogeneously fluorescent cytotoxic granules but wild-type-like killing capacity. Expression of the fluorescent fusion protein allows quantitative analyses of cytotoxic granule maturation, transport and fusion in vitro with super-resolution imaging techniques, and two-photon microscopy in living knock-ins enables the visualization of tissue rejection through individual target cell-killing events in vivo. Thus, the new mouse line is an ideal tool to study cytotoxic T lymphocyte biology and to optimize personalized immunotherapy in cancer treatment.
Collapse
Affiliation(s)
- Praneeth Chitirala
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland UniversityHomburgGermany
| | - Hsin-Fang Chang
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland UniversityHomburgGermany
| | - Paloma Martzloff
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland UniversityHomburgGermany
| | - Christiane Harenberg
- Department of Molecular Neurobiology, Max-Planck-Institute of Experimental MedicineGöttingenGermany
| | - Keerthana Ravichandran
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland UniversityHomburgGermany
| | - Midhat H Abdulreda
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of MedicineMiamiUnited States
- Department of Surgery, University of Miami Miller School of MedicineMiamiUnited States
- Department of Microbiology and Immunology, University of Miami Miller School of MedicineMiamiUnited States
- Department of Ophthalmology, University of Miami Miller School of MedicineMiamiUnited States
| | - Per-Olof Berggren
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of MedicineMiamiUnited States
- Department of Surgery, University of Miami Miller School of MedicineMiamiUnited States
- Diabetes Research Institute FederationHollywoodUnited States
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Elmar Krause
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland UniversityHomburgGermany
| | - Claudia Schirra
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland UniversityHomburgGermany
| | - Trese Leinders-Zufall
- Sensory and Neuroendocrine Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland UniversityHomburgGermany
| | - Fritz Benseler
- Department of Molecular Neurobiology, Max-Planck-Institute of Experimental MedicineGöttingenGermany
| | - Nils Brose
- Department of Molecular Neurobiology, Max-Planck-Institute of Experimental MedicineGöttingenGermany
| | - Jens Rettig
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland UniversityHomburgGermany
| |
Collapse
|
20
|
Investigation of Cytotoxic T Lymphocyte Function during Allorejection in the Anterior Chamber of the Eye. Int J Mol Sci 2020; 21:ijms21134660. [PMID: 32629968 PMCID: PMC7369940 DOI: 10.3390/ijms21134660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 01/12/2023] Open
Abstract
Cytotoxic T lymphocytes (CTL) are an essential part of our immune system by killing infected and malignant cells. To fully understand this process, it is necessary to study CTL function in the physiological setting of a living organism to account for their interplay with other immune cells like CD4+ T helper cells and macrophages. The anterior chamber of the eye (ACE), originally developed for diabetes research, is ideally suited for non-invasive and longitudinal in vivo imaging. We take advantage of the ACE window to observe immune responses, particularly allorejection of islets of Langerhans cells by CTLs. We follow the onset of the rejection after vascularization on islets until the end of the rejection process for about a month by repetitive two-photon microscopy. We find that CTLs show reduced migration on allogeneic islets in vivo compared to in vitro data, indicating CTL activation. Interestingly, the temporal infiltration pattern of T cells during rejection is precisely regulated, showing enrichment of CD4+ T helper cells on the islets before arrival of CD8+ CTLs. The adaptation of the ACE to immune responses enables the examination of the mechanism and regulation of CTL-mediated killing in vivo and to further investigate the killing in gene-deficient mice that resemble severe human immune diseases.
Collapse
|
21
|
Alcazar O, Hernandez LF, Nakayasu ES, Piehowski PD, Ansong C, Abdulreda MH, Buchwald P. Longitudinal proteomics analysis in the immediate microenvironment of islet allografts during progression of rejection. J Proteomics 2020; 223:103826. [PMID: 32442648 DOI: 10.1016/j.jprot.2020.103826] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/08/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022]
Abstract
The applicability and benefits of pancreatic islet transplantation are limited due to various issues including the need to avoid immune-mediated rejection. Here, we used our experimental platform of allogeneic islet transplant in the anterior chamber of the eye (ACE-platform) to longitudinally monitor the progress of rejection in mice and obtain aqueous humor samples representative of the microenvironment of the graft for accurately-timed proteomic analyses. LC-MS/MS-based proteomics performed on such mass-limited samples (~5 μL) identified a total of 1296 proteins. Various analyses revealed distinct protein patterns associated with the mounting of the inflammatory and immune responses and their evolution with the progression of the rejection. Pathway analyses indicated predominant changes in cytotoxic functions, cell movement, and innate and adaptive immune responses. Network prediction analyses revealed transition from humoral to cellular immune response and exacerbation of pro-inflammatory signaling. One of the proteins identified by this localized proteomics as a candidate biomarker of islet rejection, Cystatin 3, was further validated by ELISA in the aqueous humor. This study provides (1) experimental evidence demonstrating the feasibility of longitudinal localized proteomics using small aqueous humor samples and (2) proof-of-concept for the discovery of biomarkers of impending immune attack from the immediate local microenvironment of ACE-transplanted islets. SIGNIFICANCE: The combination of the ACE-platform and longitudinal localized proteomics offers a powerful approach to biomarker discovery during the various stages of immune reactions mounted against transplanted tissues including pancreatic islets. It also supports proteomics-assisted drug discovery and development efforts aimed at preventing rejection through efficacy assessment of new agents by noninvasive and longitudinal graft monitoring.
Collapse
Affiliation(s)
- Oscar Alcazar
- University of Miami Miller School of Medicine, Diabetes Research Institute, Miami, FL, USA
| | - Luis F Hernandez
- University of Miami Miller School of Medicine, Diabetes Research Institute, Miami, FL, USA
| | - Ernesto S Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Paul D Piehowski
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Charles Ansong
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Midhat H Abdulreda
- University of Miami Miller School of Medicine, Diabetes Research Institute, Miami, FL, USA; University of Miami Miller School of Medicine, Department of Surgery, Miami, FL, USA; University of Miami Miller School of Medicine, Department of Microbiology and Immunology, Miami, FL, USA; University of Miami Miller School of Medicine, Department of Ophthalmology, Miami, FL, USA.
| | - Peter Buchwald
- University of Miami Miller School of Medicine, Diabetes Research Institute, Miami, FL, USA; University of Miami Miller School of Medicine, Department of Molecular and Cellular Pharmacology, Miami, FL, USA.
| |
Collapse
|
22
|
Tun SBB, Chua M, Hasan R, Köhler M, Zheng X, Ali Y, Abdulreda MH, Juntti-Berggren L, Barathi VA, Berggren PO. Islet Transplantation to the Anterior Chamber of the Eye-A Future Treatment Option for Insulin-Deficient Type-2 Diabetics? A Case Report from a Nonhuman Type-2 Diabetic Primate. Cell Transplant 2020; 29:963689720913256. [PMID: 32264703 PMCID: PMC7444223 DOI: 10.1177/0963689720913256] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Replacement of the insulin-secreting beta cells through transplantation of pancreatic islets to the liver is a promising treatment for type-1 diabetes. However, low oxygen tension, shear stress, and the induction of inflammation lead to significant islet dysfunction and loss. The anterior chamber of the eye (ACE) has gained considerable interest and represents an alternative therapeutic islet transplantation site because of its accessibility, high oxygen tension, and immune-privileged milieu. We have previously demonstrated the feasibility of intraocular islet transplant in mouse and nonhuman primate models of type-1 diabetes and are now assessing its efficacy on glucose homeostasis in a nonhuman primate model of type-2 diabetes. We transplanted allogeneic donor islets (1,500 islet equivalents/kg) into the anterior chamber of one eye in a cynomolgus monkey with high-fat-diet-induced type-2 diabetes. Repeated examinations of the anterior and posterior segments of both eyes were done to monitor the engrafted islets and assess the overall ocular health. Fasting blood glucose level, blood biochemistry, and other metabolic parameters were routinely evaluated to determine the function of the islet graft and diabetes status. The transplanted islets were rapidly engrafted onto the iris and became vascularized 1 month after transplantation. We did not detect changes in intraocular pressure, cataract formation, ophthalmitis, or retinal vessel deformation. A significant lower fasting blood glucose level was observed while the graft was in place, and the transplantation reverts the progression of diabetes. The metabolic markers, hemoglobin A1C and fructosamine, demonstrated improvement following islet transplantation. As a conclusion, intraocular islet transplantation in one eye of a cynomolgus monkey with type-2 diabetes improved its overall plasma glucose homeostasis, as evidenced by short-term measures and long-term metabolic markers. These results further support the future application of the ACE as an alternative site for clinical islet transplants in the context of type-2 diabetes.
Collapse
Affiliation(s)
- Sai Bo Bo Tun
- Translational Pre-Clinical Model Platform, Singapore Eye Research Institute (SERI), Singapore.,The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Minni Chua
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Riasat Hasan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Martin Köhler
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Xiaofeng Zheng
- Translational Pre-Clinical Model Platform, Singapore Eye Research Institute (SERI), Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Yusuf Ali
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Midhat H Abdulreda
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lisa Juntti-Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Veluchamy A Barathi
- Translational Pre-Clinical Model Platform, Singapore Eye Research Institute (SERI), Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Ophthalmology and Visual Sciences Academic Clinical Program, DUKE-NUS Graduate Medical School, Singapore
| | - Per-Olof Berggren
- Translational Pre-Clinical Model Platform, Singapore Eye Research Institute (SERI), Singapore.,The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
23
|
Alcazar O, Hernandez LF, Tschiggfrie A, Muehlbauer MJ, Bain JR, Buchwald P, Abdulreda MH. Feasibility of Localized Metabolomics in the Study of Pancreatic Islets and Diabetes. Metabolites 2019; 9:E207. [PMID: 31569489 PMCID: PMC6835460 DOI: 10.3390/metabo9100207] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/22/2022] Open
Abstract
(1) Background: Disruption of insulin production by native or transplanted pancreatic islets caused by auto/allo-immunity leads to hyperglycemia, a serious health condition and important therapeutic challenge due to the lifelong need for exogeneous insulin administration. Early metabolic biomarkers can prompt timely interventions to preserve islet function, but reliable biomarkers are currently lacking. We explored the feasibility of "localized metabolomics" where initial biomarker discovery is made in aqueous humor samples for further validation in the circulation. (2) Methods: We conducted non-targeted metabolomic studies in parallel aqueous humor and plasma samples from diabetic and nondiabetic mice. Metabolite levels and associated pathways were compared in both compartments as well as to an earlier longitudinal dataset in hyperglycemia-progressor versus non-progressor non-obese diabetic (NOD) mice. (3) Results: We confirmed that aqueous humor samples can be used to assess metabolite levels. About half of the identified metabolites had well-correlated levels in the aqueous humor and plasma. Several plasma metabolites were significantly different between diabetic and nondiabetic animals and between males and females, and many of them were correlated with the aqueous humor. (4) Conclusions: This study provides proof-of-concept evidence that aqueous humor samples enriched with islet-related metabolites and representative of the immediate islet microenvironment following intraocular islet transplant can be used to assess metabolic changes that could otherwise be overlooked in the general circulation. The findings support localized metabolomics, with and without intraocular islet transplant, to identify biomarkers associated with diabetes and islet allograft rejection.
Collapse
Affiliation(s)
- Oscar Alcazar
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Luis F Hernandez
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Ashley Tschiggfrie
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Michael J Muehlbauer
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27708, USA.
| | - James R Bain
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27708, USA.
| | - Peter Buchwald
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Midhat H Abdulreda
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
24
|
Abdulreda MH, Molano RD, Faleo G, Lopez-Cabezas M, Shishido A, Ulissi U, Fotino C, Hernandez LF, Tschiggfrie A, Aldrich VR, Tamayo-Garcia A, Bayer AS, Ricordi C, Caicedo A, Buchwald P, Pileggi A, Berggren PO. In vivo imaging of type 1 diabetes immunopathology using eye-transplanted islets in NOD mice. Diabetologia 2019; 62:1237-1250. [PMID: 31087105 PMCID: PMC6561836 DOI: 10.1007/s00125-019-4879-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/22/2019] [Indexed: 12/31/2022]
Abstract
AIMS/HYPOTHESIS Autoimmune attack against the insulin-producing beta cells in the pancreatic islets results in type 1 diabetes. However, despite considerable research, details of the type 1 diabetes immunopathology in situ are not fully understood mainly because of difficult access to the pancreatic islets in vivo. METHODS Here, we used direct non-invasive confocal imaging of islets transplanted in the anterior chamber of the eye (ACE) to investigate the anti-islet autoimmunity in NOD mice before, during and after diabetes onset. ACE-transplanted islets allowed longitudinal studies of the autoimmune attack against islets and revealed the infiltration kinetics and in situ motility dynamics of fluorescence-labelled autoreactive T cells during diabetes development. Ex vivo immunostaining was also used to compare immune cell infiltrations into islet grafts in the eye and kidney as well as in pancreatic islets of the same diabetic NOD mice. RESULTS We found similar immune infiltration in native pancreatic and ACE-transplanted islets, which established the ACE-transplanted islets as reliable reporters of the autoimmune response. Longitudinal studies in ACE-transplanted islets identified in vivo hallmarks of islet inflammation that concurred with early immune infiltration of the islets and preceded their collapse and hyperglycaemia onset. A model incorporating data on ACE-transplanted islet degranulation and swelling allowed early prediction of the autoimmune attack in the pancreas and prompted treatments to intercept type 1 diabetes. CONCLUSIONS/INTERPRETATION The current findings highlight the value of ACE-transplanted islets in studying early type 1 diabetes pathogenesis in vivo and underscore the need for timely intervention to halt disease progression.
Collapse
Affiliation(s)
- Midhat H Abdulreda
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA.
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA.
- Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - R Damaris Molano
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
| | - Gaetano Faleo
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
| | - Maite Lopez-Cabezas
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
| | - Alexander Shishido
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
| | - Ulisse Ulissi
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
| | - Carmen Fotino
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
| | - Luis F Hernandez
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
| | - Ashley Tschiggfrie
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
| | - Virginia R Aldrich
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
| | - Alejandro Tamayo-Garcia
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Allison S Bayer
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Camillo Ricordi
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA
- Diabetes Research Institute Federation, Hollywood, FL, USA
| | - Alejandro Caicedo
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Peter Buchwald
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA.
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Antonello Pileggi
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA.
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA.
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
- Center for Scientific Review, National Institutes of Health, 6701 Rockledge Drive, Bethesda, MD, 20892, USA.
| | - Per-Olof Berggren
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA.
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
- Diabetes Research Institute Federation, Hollywood, FL, USA.
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-17176, Stockholm, Sweden.
| |
Collapse
|
25
|
Abdulreda MH, Berman DM, Shishido A, Martin C, Hossameldin M, Tschiggfrie A, Hernandez LF, Hernandez A, Ricordi C, Parel JM, Jankowska-Gan E, Burlingham WJ, Arrieta-Quintero EA, Perez VL, Kenyon NS, Berggren PO. Operational immune tolerance towards transplanted allogeneic pancreatic islets in mice and a non-human primate. Diabetologia 2019; 62:811-821. [PMID: 30701283 PMCID: PMC6451664 DOI: 10.1007/s00125-019-4814-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 12/14/2018] [Indexed: 12/31/2022]
Abstract
AIMS/HYPOTHESIS Patients with autoimmune type 1 diabetes transplanted with pancreatic islets to their liver experience significant improvement in quality of life through better control of blood sugar and enhanced awareness of hypoglycaemia. However, long-term survival and efficacy of the intrahepatic islet transplant are limited owing to liver-specific complications, such as immediate blood-mediated immune reaction, hypoxia, a highly enzymatic and inflammatory environment and locally elevated levels of drugs including immunosuppressive agents, all of which are injurious to islets. This has spurred a search for new islet transplant sites and for innovative ways to achieve long-term graft survival and efficacy without life-long systemic immunosuppression and its complications. METHODS We used our previously established approach of islet transplant in the anterior chamber of the eye in allogeneic recipient mouse models and a baboon model of diabetes, which were treated transiently with anti-CD154/CD40L blocking antibody in the peri-transplant period. Survival of the intraocular islet allografts was assessed by direct visualisation in the eye and metabolic variables (blood glucose and C-peptide measurements). We evaluated longitudinally the cytokine profile in the local microenvironment of the intraocular islet allografts, represented in aqueous humour, under conditions of immune rejection vs tolerance. We also evaluated the recall response in the periphery of the baboon recipient using delayed-type hypersensitivity (DTH) assay, and in mice after repeat transplant in the kidney following initial transplant with allogeneic islets in the eye or kidney. RESULTS Results in mice showed >300 days immunosuppression-free survival of allogeneic islets transplanted in the eye or kidney. Notably, >70% of tolerant mice, initially transplanted in the eye, exhibited >400 days of graft survival after re-transplant in the kidney without immunosuppression compared with ~30% in mice that were initially transplanted in the kidney. Cytokine and DTH data provided evidence of T helper 2-driven local and peripheral immune regulatory mechanisms in support of operational immune tolerance towards the islet allografts in both models. CONCLUSIONS/INTERPRETATION We are currently evaluating the safety and efficacy of intraocular islet transplantation in a phase 1 clinical trial. In this study, we demonstrate immunosuppression-free long-term survival of intraocular islet allografts in mice and in a baboon using transient peri-transplant immune intervention. These results highlight the potential for inducing islet transplant immune tolerance through the intraocular route. Therefore, the current findings are conceptually significant and may impact markedly on clinical islet transplantation in the treatment of diabetes.
Collapse
Affiliation(s)
- Midhat H Abdulreda
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA.
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA.
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Dora M Berman
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alexander Shishido
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
| | - Christopher Martin
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
| | - Maged Hossameldin
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
| | - Ashley Tschiggfrie
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
| | - Luis F Hernandez
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
| | - Ana Hernandez
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
| | - Camillo Ricordi
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Diabetes Research Institute Federation, Hollywood, FL, USA
| | - Jean-Marie Parel
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ewa Jankowska-Gan
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - William J Burlingham
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | | | - Victor L Perez
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Duke Ophthalmology, Duke University, Durham, NC, USA
| | - Norma S Kenyon
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Per-Olof Berggren
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA.
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-17176, Stockholm, Sweden.
| |
Collapse
|
26
|
Local release of rapamycin by microparticles delays islet rejection within the anterior chamber of the eye. Sci Rep 2019; 9:3918. [PMID: 30850640 PMCID: PMC6408557 DOI: 10.1038/s41598-019-40404-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/14/2019] [Indexed: 12/23/2022] Open
Abstract
The anterior chamber of the eye (ACE) has emerged as a promising clinical islet transplantation site because of its multiple advantages over the conventional intra-hepatic portal site. This includes reduced surgical invasiveness and increased islet graft survival rate. It also allows for enhanced accessibility and monitoring of the islets. Although the ACE is initially an immuno-privileged site, this privilege is disrupted once the islet grafts are re-vascularized. Given that the ACE is a confined space, achieving graft immune tolerance through local immunosuppressive drug delivery is therefore feasible. Here, we show that islet rejection in the ACE of mice can be significantly suppressed through local delivery of rapamycin by carefully designed sustained-release microparticles. In this 30-day study, allogeneic islet grafts with blank microparticles were completely rejected 18 days post-transplantation into mice. Importantly, allogeneic islet grafts co-injected with rapamycin releasing microparticles into a different eye of the same recipient were preserved much longer, with some grafts surviving for more than 30 days. Hence, islet allograft survival was enhanced by a localized and prolonged delivery of an immunosuppressive drug. We envisage that this procedure will relieve diabetic transplant recipients from harsh systemic immune suppression, while achieving improved glycemic control and reduced insulin dependence.
Collapse
|
27
|
Abstract
Diabetes develops due to deficient functional β cell mass, insulin resistance, or both. Yet, various challenges in understanding the mechanisms underlying diabetes development in vivo remain to be overcome owing to the lack of appropriate intravital imaging technologies. To meet these challenges, we have exploited the anterior chamber of the eye (ACE) as a novel imaging site to understand diabetes basics and clinics in vivo. We have developed a technology platform transplanting pancreatic islets into the ACE where they later on can be imaged non-invasively for long time. It turns out that the ACE serves as an optimal imaging site and provides implanted islets with an oxygen-rich milieu and an immune-privileged niche where they undergo optimal engraftment, rich vascularization and dense innervation, preserve organotypic features and live with satisfactory viability and functionality. The ACE technology has led to a series of significant observations. It enables in vivo microscopy of islet cytoarchitecture, function and viability in the physiological context and intravital imaging of a variety of pathological events such as autoimmune insulitis, defects in β cell function and mass and insulin resistance during diabetes development in a real-time manner. Furthermore, application of the ACE technology in humanized mice and non-human primates verifies translational and clinical values of the technology. In this article, we describe the ACE technology in detail, review accumulated knowledge gained by means of the ACE technology and delineate prospective avenues for the ACE technology.
Collapse
|
28
|
Yang J, Park JW, Zheng D, Xu RH. Universal Corneal Epithelial-Like Cells Derived from Human Embryonic Stem Cells for Cellularization of a Corneal Scaffold. Transl Vis Sci Technol 2018; 7:23. [PMID: 30323996 PMCID: PMC6181193 DOI: 10.1167/tvst.7.5.23] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/07/2018] [Indexed: 12/13/2022] Open
Abstract
Purpose We generated universal corneal epithelial cells (CEC) from human embryonic stem cells (hESC) by genetically removing human leukocyte antigens (HLA) class I from the cell surface. Methods The serum-free, growth factor-free, and defined medium E6 was used to differentiate hESC to CEC. Decellularized murine corneas were recellularized with hESC-derived CEC. Using CRISPR/Cas9, β-2-microglobulin (B2M) was deleted in hESC to block the assembly of HLA class-I antigens on the cell surface to generate B2M−/− CEC. Results E6 alone was sufficient to allow hESC differentiation to CEC. A time-course analysis of the global gene expression of the differentiating cells indicates that the differentiation closely resembles the corneal development in vivo. The hESC-CEC were highly proliferative, and could form multilayer epithelium in decellularized murine cornea, retain its transparency, and form intact tight junctions on its surface. As reported before, B2M knockout led to the absence of HLA class-I on the cell surface of hESC and subsequently derived CEC following stimulation with inflammatory factors. Moreover, B2M−/− CEC, following transplantation into mouse eyes, caused less T-cell infiltration in the limbal region of the eye than the wild-type control. Conclusions CEC can be derived from hESC via a novel and simple protocol free of any proteins, hESC-CEC seeded on decellularized animal cornea form tight junctions and allow light transmittance, and B2M−/− CEC are hypoimmunogenic both in vitro and in vivo. Translational Relevance B2M−/− hESC-CEC can be an unlimited and universal therapy for corneal repair in patients of any HLA type.
Collapse
Affiliation(s)
- Juan Yang
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Jung Woo Park
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Dejin Zheng
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Ren-He Xu
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
29
|
Oltra E, Caicedo A. Real Time In Vivo Tracking of Thymocytes in the Anterior Chamber of the Eye by Laser Scanning Microscopy. J Vis Exp 2018. [PMID: 30346412 DOI: 10.3791/58236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The purpose of the method being presented is to show, for the first time, the transplant of newborn thymi into the anterior eye chamber of isogenic adult mice for in vivo longitudinal real-time monitoring of thymocytes´ dynamics within a vascularized thymus segment. Following the transplantation, laser scanning microscopy (LSM) through the cornea allows in vivo noninvasive repeated imaging at cellular resolution level. Importantly, the approach adds to previous intravital T-cell maturation imaging models the possibility for continuous progenitor cell recruitment and mature T-cell egress recordings in the same animal. Additional advantages of the system are the transparency of the grafted area, permitting macroscopic rapid monitoring of the implanted tissue, and the accessibility to the implant allowing for localized in addition to systemic treatments. The main limitation being the volume of the tissue that fits in the reduced space of the eye chamber which demands for lobe trimming. Organ integrity is maximized by dissecting thymus lobes in patterns previously shown to be functional for mature T-cell production. The technique is potentially suited to interrogate a milieu of medically relevant questions related to thymus function that include autoimmunity, immunodeficiency and central tolerance; processes which remain mechanistically poorly defined. The fine dissection of mechanisms guiding thymocyte migration, differentiation and selection should lead to novel therapeutic strategies targeting developing T cells.
Collapse
Affiliation(s)
- Elisa Oltra
- School of Medicine and Dentistry, Universidad Católica de Valencia San Vicente Mártir; Unidad Mixta CIPF-UCV, Centro de Investigación Príncipe Felipe;
| | | |
Collapse
|
30
|
Paschen M, Moede T, Valladolid-Acebes I, Leibiger B, Moruzzi N, Jacob S, García-Prieto CF, Brismar K, Leibiger IB, Berggren PO. Diet-induced β-cell insulin resistance results in reversible loss of functional β-cell mass. FASEB J 2018; 33:204-218. [PMID: 29957055 PMCID: PMC6355083 DOI: 10.1096/fj.201800826r] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although convincing in genetic models, the relevance of β-cell insulin resistance in diet-induced type 2 diabetes (T2DM) remains unclear. Exemplified by diabetes-prone, male, C57B1/6J mice being fed different combinations of Western-style diet, we show that β-cell insulin resistance occurs early during T2DM progression and is due to a combination of lipotoxicity and increased β-cell workload. Within 8 wk of being fed a high-fat, high-sucrose diet, mice became obese, developed impaired insulin and glucose tolerances, and displayed noncompensatory insulin release, due, at least in part, to reduced expression of syntaxin-1A. Through reporter islets transplanted to the anterior chamber of the eye, we demonstrated a concomitant loss of functional β-cell mass. When mice were changed from diabetogenic diet to normal chow diet, the diabetes phenotype was reversed, suggesting a remarkable plasticity of functional β-cell mass in the early phase of T2DM development. Our data reinforce the relevance of diet composition as an environmental factor determining different routes of diabetes progression in a given genetic background. Employing the in vivo reporter islet–monitoring approach will allow researchers to define key times in the dynamics of reversible loss of functional β-cell mass and, thus, to investigate the underlying, molecular mechanisms involved in the progression toward T2DM manifestation.—Paschen, M., Moede, T., Valladolid-Acebes, I., Leibiger, B., Moruzzi, N., Jacob, S., García-Prieto, C. F., Brismar, K., Leibiger, I. B., Berggren, P.-O. Diet-induced β-cell insulin resistance results in reversible loss of functional β-cell mass.
Collapse
Affiliation(s)
- Meike Paschen
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Tilo Moede
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Ismael Valladolid-Acebes
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Barbara Leibiger
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Noah Moruzzi
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Stefan Jacob
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Concha F García-Prieto
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Kerstin Brismar
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Ingo B Leibiger
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
31
|
Rodriguez-Diaz R, Molano RD, Weitz JR, Abdulreda MH, Berman DM, Leibiger B, Leibiger IB, Kenyon NS, Ricordi C, Pileggi A, Caicedo A, Berggren PO. Paracrine Interactions within the Pancreatic Islet Determine the Glycemic Set Point. Cell Metab 2018; 27. [PMID: 29514065 PMCID: PMC5872154 DOI: 10.1016/j.cmet.2018.01.015] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Every animal species has a signature blood glucose level or glycemic set point. These set points are different, and the normal glycemic levels (normoglycemia) of one species would be life threatening for other species. Mouse normoglycemia can be considered diabetic for humans. The biological determinants of the glycemic set point remain unclear. Here we show that the pancreatic islet imposes its glycemic set point on the organism, making it the bona fide glucostat in the body. Moreover, and in contrast to rodent islets, glucagon input from the alpha cell to the insulin-secreting beta cell is necessary to fine-tune the distinctive human set point. These findings affect transplantation and regenerative approaches to treat diabetes because restoring normoglycemia may require more than replacing only the beta cells. Furthermore, therapeutic strategies using glucagon receptor antagonists as hypoglycemic agents need to be reassessed, as they may reset the overall glucostat in the organism.
Collapse
Affiliation(s)
- Rayner Rodriguez-Diaz
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10(th) Avenue, Miami, FL 33136, USA; Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm 17177, Sweden.
| | - R Damaris Molano
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jonathan R Weitz
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10(th) Avenue, Miami, FL 33136, USA
| | - Midhat H Abdulreda
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Dora M Berman
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Barbara Leibiger
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Ingo B Leibiger
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Norma S Kenyon
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Antonello Pileggi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alejandro Caicedo
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10(th) Avenue, Miami, FL 33136, USA; Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Program in Neuroscience, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| | - Per-Olof Berggren
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm 17177, Sweden; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore; Pancreatic Islet Biology and Diabetes Consortium, Imperial College, London, UK.
| |
Collapse
|
32
|
Mohan JF, Kohler RH, Hill JA, Weissleder R, Mathis D, Benoist C. Imaging the emergence and natural progression of spontaneous autoimmune diabetes. Proc Natl Acad Sci U S A 2017; 114:E7776-E7785. [PMID: 28839093 PMCID: PMC5604023 DOI: 10.1073/pnas.1707381114] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Type 1 diabetes in the nonobese diabetic mouse stems from an infiltration of the pancreatic islets by a mixed population of immunocytes, which results in the impairment and eventual destruction of insulin-producing β-cells. Little is known about the dynamics of lymphocyte movement in the pancreas during disease progression. Using advanced intravital imaging approaches and newly created reporter mice (Flt3-BFP2, Mertk-GFP-DTR, Cd4-tdTomato, Cd8a-tdTomato), we show that the autoimmune process initiates first with a T cell infiltration into the islets, where they have restricted mobility but reside and are activated in apposition to CX3CR1+ macrophages. The main expansion then occurs in the connective tissue outside the islet, which remains more or less intact. CD4+ and CD8+ T cells, Tregs, and dendritic cells (DCs) are highly mobile, going along microvascular tracks, while static macrophages (MF) form a more rigid structure, often encasing the islet cell mass. Transient cell-cell interactions are formed between T cells and both MFs and DCs, but also surprisingly between MFs and DCs themselves, possibly denoting antigen transfer. In later stages, extensive islet destruction coincides with preferential antigen presentation to, and activation of, CD8+ T cells. Throughout the process, Tregs patrol the active compartments, consistent with the notion that they control the activation of many cell types.
Collapse
Affiliation(s)
- James F Mohan
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Rainer H Kohler
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114
| | - Jonathan A Hill
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114;
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115
| | - Diane Mathis
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115;
| | - Christophe Benoist
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115;
| |
Collapse
|
33
|
Leibiger IB, Berggren PO. Intraocular in vivo imaging of pancreatic islet cell physiology/pathology. Mol Metab 2017; 6:1002-1009. [PMID: 28951824 PMCID: PMC5605725 DOI: 10.1016/j.molmet.2017.03.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/07/2017] [Accepted: 03/18/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Diabetes mellitus has reached epidemic proportions and requires new strategies for treatment. Unfortunately, the efficacy of treatment regimens on maintaining/re-gaining functional beta cell mass can, at the present, only be determined indirectly. Direct monitoring of beta cell mass is complicated by the anatomy of the endocrine pancreas, which consists of thousands to a million of discrete micro-organs, i.e. islets of Langerhans, which are scattered throughout the pancreas. SCOPE OF REVIEW Here, we review the progress made over the last years using the anterior chamber of the eye as a transplantation site for functional imaging of pancreatic islet cells in the living organism. Islets engrafted on the iris are vascularized and innervated and the cornea, serving as a natural body-window, allows for microscopic, non-invasive, longitudinal evaluation of islet/beta cell function and survival with single-cell resolution in health and disease. MAJOR CONCLUSIONS Data provided by us and others demonstrate the high versatility of this imaging platform. The use of 'reporter islets' engrafted in the eye, reporting on the status of in situ endogenous islets in the pancreas of the same animal, allows the identification of key-events in the development and progression of diabetes. This will not only serve as a versatile research tool but will also lay the foundation for a personalized medicine approach and will serve as a screening platform for new drugs and/or treatment protocols. 'Metabolic' islet transplantation, in which islets engrafted in the eye replace the endogenous beta cells, will allow for the establishment of islet-specific transgenic models and 'humanized' mouse models as well as serving as the basis for a new clinical transplantation site for the cure of diabetes.
Collapse
Affiliation(s)
- Ingo B. Leibiger
- The Rolf Luft Research Center for Diabetes and Endocrinology, L1:03 Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, L1:03 Karolinska Institutet, SE-171 76 Stockholm, Sweden
| |
Collapse
|
34
|
Hsueh B, Burns VM, Pauerstein P, Holzem K, Ye L, Engberg K, Wang AC, Gu X, Chakravarthy H, Arda HE, Charville G, Vogel H, Efimov IR, Kim S, Deisseroth K. Pathways to clinical CLARITY: volumetric analysis of irregular, soft, and heterogeneous tissues in development and disease. Sci Rep 2017; 7:5899. [PMID: 28724969 PMCID: PMC5517617 DOI: 10.1038/s41598-017-05614-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/12/2017] [Indexed: 12/24/2022] Open
Abstract
Three-dimensional tissue-structural relationships are not well captured by typical thin-section histology, posing challenges for the study of tissue physiology and pathology. Moreover, while recent progress has been made with intact methods for clearing, labeling, and imaging whole organs such as the mature brain, these approaches are generally unsuitable for soft, irregular, and heterogeneous tissues that account for the vast majority of clinical samples and biopsies. Here we develop a biphasic hydrogel methodology, which along with automated analysis, provides for high-throughput quantitative volumetric interrogation of spatially-irregular and friable tissue structures. We validate and apply this approach in the examination of a variety of developing and diseased tissues, with specific focus on the dynamics of normal and pathological pancreatic innervation and development, including in clinical samples. Quantitative advantages of the intact-tissue approach were demonstrated compared to conventional thin-section histology, pointing to broad applications in both research and clinical settings.
Collapse
Affiliation(s)
- Brian Hsueh
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Vanessa M Burns
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, 94305, USA
| | - Philip Pauerstein
- Department of Developmental Biology, Stanford University, Stanford, CA, 94305, USA
| | - Katherine Holzem
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Li Ye
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
- Howard Hughes Medical Institute, Stanford, CA, 94305, USA
| | - Kristin Engberg
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Ai-Chi Wang
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Xueying Gu
- Department of Developmental Biology, Stanford University, Stanford, CA, 94305, USA
| | - Harini Chakravarthy
- Department of Developmental Biology, Stanford University, Stanford, CA, 94305, USA
| | - H Efsun Arda
- Department of Developmental Biology, Stanford University, Stanford, CA, 94305, USA
| | - Gregory Charville
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Hannes Vogel
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Igor R Efimov
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA
- Department of Biomedical Engineering, The George Washington University, Washington, DC, 20052, USA
| | - Seung Kim
- Department of Developmental Biology, Stanford University, Stanford, CA, 94305, USA
- Howard Hughes Medical Institute, Stanford, CA, 94305, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA.
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA.
- Howard Hughes Medical Institute, Stanford, CA, 94305, USA.
| |
Collapse
|
35
|
Assessment of Immune Isolation of Allogeneic Mouse Pancreatic Progenitor Cells by a Macroencapsulation Device. Transplantation 2017; 100:1211-8. [PMID: 26982952 DOI: 10.1097/tp.0000000000001146] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Embryonic stem cell (ESC)-derived β cells hold the promise of providing a renewable source of tissue for the treatment of insulin-dependent diabetes. Encapsulation may allow ESC-derived β cells to be transplanted without immunosuppression, thus enabling wider application of this therapy. METHODS In this study, we investigated the immunogenicity of mouse pancreatic progenitor cells and efficacy of a new macroencapsulation device in protecting these cells against alloimmune and autoimmune responses in mouse models. RESULTS Mouse pancreatic progenitor cells activated the indirect but not the direct pathway of alloimmune response and were promptly rejected in immune competent hosts. The new macroencapsulation device abolished T cell activation induced by allogeneic splenocytes and protected allogeneic MIN6 β cells and pancreatic progenitors from rejection even in presensitized recipients. In addition, the device was effective in protecting MIN6 cells in spontaneously diabetic nonobese diabetic recipients against both alloimmune and recurring autoimmune responses. CONCLUSIONS Our results demonstrate that macroencapsulation can effectively prevent immune sensing and rejection of allogeneic pancreatic progenitor cells in fully sensitized and autoimmune hosts.
Collapse
|
36
|
Park S, Lee DY. The anterior chamber of the eye as a site for pancreatic islet transplantation. J IND ENG CHEM 2017. [DOI: 10.1016/j.jiec.2017.02.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
37
|
Shishido A, Caicedo A, Rodriguez-Diaz R, Pileggi A, Berggren PO, Abdulreda MH. Clinical intraocular islet transplantation is not a number issue. CELLR4-- REPAIR, REPLACEMENT, REGENERATION, & REPROGRAMMING 2016; 4:e2120. [PMID: 29497631 PMCID: PMC5828508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
It is now well established that beta cell replacement through pancreatic islet transplantation results in significant improvement in the quality-of-life of type 1 diabetes (T1D) patients. This is achieved through improved control and prevention of severe drops in blood sugar levels. Islet transplant therapy is on the verge of becoming standard-of-care in the USA. Yet, as with other established transplantation therapies, there remain hurdles to overcome to bring islet transplantation to full fruition as a long-lasting therapy of T1D. One of these hurdles is establishing reliable new sites, other than the liver, where durable efficacy and survival of transplanted islets can be achieved. In this article, we discuss the anterior chamber of the eye as a new site for clinical islet transplantation in the treatment of T1D. We specifically focus on the common conceptions, and preconceptions, on the requirements of islet mass, and whether or not the anterior chamber can accommodate sufficient islets to achieve meaningful efficacy and significant impact on hyperglycemia in clinical application.
Collapse
Affiliation(s)
- A Shishido
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - A Caicedo
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, Division of Endocrinology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - R Rodriguez-Diaz
- Department of Medicine, Division of Endocrinology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - A Pileggi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Departments of Microbiology and Immunology and Biomedical Engineering, University of Miami, Miami, FL, USA
| | - P-O Berggren
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - M H Abdulreda
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
38
|
Boden J, Lassance-Soares RM, Wang H, Wei Y, Spiga MG, Adi J, Layman H, Yu H, Vazquez-Padron RI, Andreopoulos F, Webster KA. Vascular Regeneration in Ischemic Hindlimb by Adeno-Associated Virus Expressing Conditionally Silenced Vascular Endothelial Growth Factor. J Am Heart Assoc 2016; 5:e001815. [PMID: 27231018 PMCID: PMC4937238 DOI: 10.1161/jaha.115.001815] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 04/19/2016] [Indexed: 01/07/2023]
Abstract
BACKGROUND Critical limb ischemia (CLI) is the extreme manifestation of peripheral artery disease, a major unmet clinical need for which lower limb amputation is the only option for many patients. After 2 decades in development, therapeutic angiogenesis has been tested clinically via intramuscular delivery of proangiogenic proteins, genes, and stem cells. Efficacy has been modest to absent, and the largest phase 3 trial of gene therapy for CLI reported a worsening trend of plasmid fibroblast growth factor. In all clinical trials to date, gene therapy has used unregulated vectors with limited duration of expression. Only unregulated extended expression vectors such as adeno-associated virus (AAV) and lentivirus have been tested in preclinical models. METHODS AND RESULTS We present preclinical results of ischemia (hypoxia)-regulated conditionally silenced (CS) AAV-human vascular endothelial growth factor (hVEGF) gene delivery that shows efficacy and safety in a setting where other strategies fail. In a BALB/c mouse model of CLI, we show that gene therapy with AAV-CS-hVEGF, but not unregulated AAV or plasmid, vectors conferred limb salvage, protection from necrosis, and vascular regeneration when delivered via intramuscular or intra-arterial routes. All vector treatments conferred increased capillary density, but organized longitudinal arteries were selectively generated by AAV-CS-hVEGF. AAV-CS-hVEGF therapy reversibly activated angiogenic and vasculogenic genes, including Notch, SDF1, Angiopoietin, and Ephrin-B2. Reoxygenation extinguished VEGF expression and inactivated the program with no apparent adverse side effects. CONCLUSIONS Restriction of angiogenic growth factor expression to regions of ischemia supports the safe and stable reperfusion of hindlimbs in a clinically relevant murine model of CLI.
Collapse
Affiliation(s)
- Jeffrey Boden
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Roberta Marques Lassance-Soares
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Huilan Wang
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Yuntao Wei
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL
| | - Maria-Grazia Spiga
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL
| | - Jennipher Adi
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL
| | - Hans Layman
- Department of Bioengineering, University of Miami Miller School of Medicine, Miami, FL
| | - Hong Yu
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Roberto I Vazquez-Padron
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Fotios Andreopoulos
- Department of Bioengineering, University of Miami Miller School of Medicine, Miami, FL
| | - Keith A Webster
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
39
|
Liu ZJ, Daftarian P, Kovalski L, Wang B, Tian R, Castilla DM, Dikici E, Perez VL, Deo S, Daunert S, Velazquez OC. Directing and Potentiating Stem Cell-Mediated Angiogenesis and Tissue Repair by Cell Surface E-Selectin Coating. PLoS One 2016; 11:e0154053. [PMID: 27104647 PMCID: PMC4841581 DOI: 10.1371/journal.pone.0154053] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 04/07/2016] [Indexed: 01/12/2023] Open
Abstract
Stem cell therapy has emerged as a promising approach for treatment of a number of diseases, including delayed and non-healing wounds. However, targeted systemic delivery of therapeutic cells to the dysfunctional tissues remains one formidable challenge. Herein, we present a targeted nanocarrier-mediated cell delivery method by coating the surface of the cell to be delivered with dendrimer nanocarriers modified with adhesion molecules. Infused nanocarrier-coated cells reach to destination via recognition and association with the counterpart adhesion molecules highly or selectively expressed on the activated endothelium in diseased tissues. Once anchored on the activated endothelium, nanocarriers-coated transporting cells undergo transendothelial migration, extravasation and homing to the targeted tissues to execute their therapeutic role. We now demonstrate feasibility, efficacy and safety of our targeted nanocarrier for delivery of bone marrow cells (BMC) to cutaneous wound tissues and grafted corneas and its advantages over conventional BMC transplantation in mouse models for wound healing and neovascularization. This versatile platform is suited for targeted systemic delivery of virtually any type of therapeutic cell.
Collapse
Affiliation(s)
- Zhao-Jun Liu
- Department of Surgery, School of Medicine, University of Miami, Coral Gables, Florida, 33136, United States of America
| | - Pirouz Daftarian
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Miami, Coral Gables, Florida, 33136, United States of America
- Dr. JT Macdonald Biomedical Nanotechnology Institute, University of Miami, Coral Gables, Florida, 33136, United States of America
| | - Letícia Kovalski
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Miami, Coral Gables, Florida, 33136, United States of America
| | - Bo Wang
- Department of Surgery, School of Medicine, University of Miami, Coral Gables, Florida, 33136, United States of America
| | - Runxia Tian
- Department of Surgery, School of Medicine, University of Miami, Coral Gables, Florida, 33136, United States of America
| | - Diego M. Castilla
- Department of Surgery, School of Medicine, University of Miami, Coral Gables, Florida, 33136, United States of America
| | - Emre Dikici
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Miami, Coral Gables, Florida, 33136, United States of America
| | - Victor L. Perez
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami, Coral Gables, Florida, 33136, United States of America
| | - Sapna Deo
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Miami, Coral Gables, Florida, 33136, United States of America
- Dr. JT Macdonald Biomedical Nanotechnology Institute, University of Miami, Coral Gables, Florida, 33136, United States of America
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Miami, Coral Gables, Florida, 33136, United States of America
- Dr. JT Macdonald Biomedical Nanotechnology Institute, University of Miami, Coral Gables, Florida, 33136, United States of America
- * E-mail: (OV); (SD)
| | - Omaida C. Velazquez
- Department of Surgery, School of Medicine, University of Miami, Coral Gables, Florida, 33136, United States of America
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Miami, Coral Gables, Florida, 33136, United States of America
- * E-mail: (OV); (SD)
| |
Collapse
|
40
|
Zattara EE, Turlington KW, Bely AE. Long-term time-lapse live imaging reveals extensive cell migration during annelid regeneration. BMC DEVELOPMENTAL BIOLOGY 2016; 16:6. [PMID: 27006129 PMCID: PMC4804569 DOI: 10.1186/s12861-016-0104-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/10/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND Time-lapse imaging has proven highly valuable for studying development, yielding data of much finer resolution than traditional "still-shot" studies and allowing direct examination of tissue and cell dynamics. A major challenge for time-lapse imaging of animals is keeping specimens immobile yet healthy for extended periods of time. Although this is often feasible for embryos, the difficulty of immobilizing typically motile juvenile and adult stages remains a persistent obstacle to time-lapse imaging of post-embryonic development. RESULTS Here we describe a new method for long-duration time-lapse imaging of adults of the small freshwater annelid Pristina leidyi and use this method to investigate its regenerative processes. Specimens are immobilized with tetrodotoxin, resulting in irreversible paralysis yet apparently normal regeneration, and mounted in agarose surrounded by culture water or halocarbon oil, to prevent dehydration but allowing gas exchange. Using this method, worms can be imaged continuously and at high spatial-temporal resolution for up to 5 days, spanning the entire regeneration process. We performed a fine-scale analysis of regeneration growth rate and characterized cell migration dynamics during early regeneration. Our studies reveal the migration of several putative cell types, including one strongly resembling published descriptions of annelid neoblasts, a cell type suggested to be migratory based on "still-shot" studies and long hypothesized to be linked to regenerative success in annelids. CONCLUSIONS Combining neurotoxin-based paralysis, live mounting techniques and a starvation-tolerant study system has allowed us to obtain the most extensive high-resolution longitudinal recordings of full anterior and posterior regeneration in an invertebrate, and to detect and characterize several cell types undergoing extensive migration during this process. We expect the tetrodotoxin paralysis and time-lapse imaging methods presented here to be broadly useful in studying other animals and of particular value for studying post-embryonic development.
Collapse
Affiliation(s)
- Eduardo E. Zattara
- Department of Biology, University of Maryland, College Park, MD 20740 USA
| | - Kate W. Turlington
- Department of Biology, University of Maryland, College Park, MD 20740 USA
| | - Alexandra E. Bely
- Department of Biology, University of Maryland, College Park, MD 20740 USA
| |
Collapse
|
41
|
Galanzha EI, Viegas MG, Malinsky TI, Melerzanov AV, Juratli MA, Sarimollaoglu M, Nedosekin DA, Zharov VP. In vivo acoustic and photoacoustic focusing of circulating cells. Sci Rep 2016; 6:21531. [PMID: 26979811 PMCID: PMC4793240 DOI: 10.1038/srep21531] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/04/2016] [Indexed: 01/21/2023] Open
Abstract
In vivo flow cytometry using vessels as natural tubes with native cell flows has revolutionized the study of rare circulating tumor cells in a complex blood background. However, the presence of many blood cells in the detection volume makes it difficult to count each cell in this volume. We introduce method for manipulation of circulating cells in vivo with the use of gradient acoustic forces induced by ultrasound and photoacoustic waves. In a murine model, we demonstrated cell trapping, redirecting and focusing in blood and lymph flow into a tight stream, noninvasive wall-free transportation of blood, and the potential for photoacoustic detection of sickle cells without labeling and of leukocytes targeted by functionalized nanoparticles. Integration of cell focusing with intravital imaging methods may provide a versatile biological tool for single-cell analysis in circulation, with a focus on in vivo needleless blood tests, and preclinical studies of human diseases in animal models.
Collapse
Affiliation(s)
- Ekaterina I Galanzha
- Arkansas Nanomedicine Center, University of Arkansas for Medical Sciences (UAMS), Little Rock, Arkansas 72205
| | - Mark G Viegas
- Arkansas Nanomedicine Center, University of Arkansas for Medical Sciences (UAMS), Little Rock, Arkansas 72205
| | - Taras I Malinsky
- Bauman Moscow State Technical University, Moscow, Russia, 107005
| | | | - Mazen A Juratli
- Arkansas Nanomedicine Center, University of Arkansas for Medical Sciences (UAMS), Little Rock, Arkansas 72205
| | - Mustafa Sarimollaoglu
- Arkansas Nanomedicine Center, University of Arkansas for Medical Sciences (UAMS), Little Rock, Arkansas 72205
| | - Dmitry A Nedosekin
- Arkansas Nanomedicine Center, University of Arkansas for Medical Sciences (UAMS), Little Rock, Arkansas 72205
| | - Vladimir P Zharov
- Arkansas Nanomedicine Center, University of Arkansas for Medical Sciences (UAMS), Little Rock, Arkansas 72205.,Moscow Institute of Physics and Technology (MIPT), Moscow Region, 141700, Russia
| |
Collapse
|
42
|
Abdulreda MH, Rodriguez-Diaz R, Caicedo A, Berggren PO. Liraglutide Compromises Pancreatic β Cell Function in a Humanized Mouse Model. Cell Metab 2016; 23:541-6. [PMID: 26876561 PMCID: PMC4785083 DOI: 10.1016/j.cmet.2016.01.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/30/2015] [Accepted: 01/15/2016] [Indexed: 01/29/2023]
Abstract
Incretin mimetics are frequently used in the treatment of type 2 diabetes because they potentiate β cell response to glucose. Clinical evidence showing short-term benefits of such therapeutics (e.g., liraglutide) is abundant; however, there have been several recent reports of unexpected complications in association with incretin mimetic therapy. Importantly, clinical evidence on the potential effects of such agents on the β cell and islet function during long-term, multiyear use remains lacking. We now show that prolonged daily liraglutide treatment of >200 days in humanized mice, transplanted with human pancreatic islets in the anterior chamber of the eye, is associated with compromised release of human insulin and deranged overall glucose homeostasis. These findings raise concern about the chronic potentiation of β cell function through incretin mimetic therapy in diabetes.
Collapse
Affiliation(s)
- Midhat H Abdulreda
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, FL 33136, USA.
| | - Rayner Rodriguez-Diaz
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, Miami, FL 33136, USA
| | - Alejandro Caicedo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, Miami, FL 33136, USA
| | - Per-Olof Berggren
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, FL 33136, USA; The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm SE-17176, Sweden.
| |
Collapse
|
43
|
Cras-Méneur C, Elghazi L, Fort P, Bernal-Mizrachi E. Noninvasive in vivo imaging of embryonic β-cell development in the anterior chamber of the eye. Islets 2016; 8:35-47. [PMID: 26950054 PMCID: PMC4878273 DOI: 10.1080/19382014.2016.1148236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
The fetal environment plays a decisive role in modifying the risk for developing diabetes later in life. Developing novel methodology for noninvasive imaging of β-cell development in vivo under the controlled physiological conditions of the host can serve to understand how this environment affects β-cell growth and differentiation. A number of culture models have been designed for pancreatic rudiment but none match the complexity of the in utero or even normal physiological environment. Speier et al. recently developed a platform of noninvasive in vivo imaging of pancreatic islets using the anterior chamber of the eye where islets get vascularized, grow and respond to physiological changes. The same methodology was adapted for the study of pancreatic development. E13.0, still undifferentiated rudiments with fluorescent lineage tracing were implanted in the AC of the eye, allowing the longitudinal study of their growth and differentiation. Within 48 h the anlages get vascularized and grow but their mesenchyme displays a selective growth advantage. The resulting imbalance leads to alteration in the differentiation pattern of the progenitors. Reducing the mesenchyme to its bare minimum before implantation allows the restoration of a proper balance and a development that mimics the normal pancreatic development. These groundbreaking observations demonstrate that the anterior chamber of the eye provides a good system for noninvasive in vivo fluorescence imaging of the developing pancreas under the physiology of the host and can have important implications for designing strategies to prevent or reverse the deleterious effects of hyperglycemia on altering β-cell function later in life.
Collapse
Affiliation(s)
- Corentin Cras-Méneur
- Internal Medicine Department, Division of Metabolism, Endocrinology and Diabetes, University of Michigan in Ann Arbor, Ann Arbor, Michigan, USA
| | - Lynda Elghazi
- Internal Medicine Department, Division of Metabolism, Endocrinology and Diabetes, University of Michigan in Ann Arbor, Ann Arbor, Michigan, USA
| | - Patrice Fort
- Ophthalmology Department, University of Michigan in Ann Arbor, Ann Arbor, Michigan, USA
| | - Ernesto Bernal-Mizrachi
- Internal Medicine Department, Division of Metabolism, Endocrinology and Diabetes, University of Michigan in Ann Arbor, Ann Arbor, Michigan, USA
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| |
Collapse
|
44
|
Berclaz C, Schmidt-Christensen A, Szlag D, Extermann J, Hansen L, Bouwens A, Villiger M, Goulley J, Schuit F, Grapin-Botton A, Lasser T, Holmberg D. Longitudinal three-dimensional visualisation of autoimmune diabetes by functional optical coherence imaging. Diabetologia 2016; 59:550-9. [PMID: 26613896 DOI: 10.1007/s00125-015-3819-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 10/30/2015] [Indexed: 10/22/2022]
Abstract
AIMS/HYPOTHESIS It is generally accepted that structural and functional quantitative imaging of individual islets would be beneficial to elucidate the pathogenesis of type 1 diabetes. We here introduce functional optical coherence imaging (FOCI) for fast, label-free monitoring of beta cell destruction and associated alterations of islet vascularisation. METHODS NOD mouse and human islets transplanted into the anterior chamber of the eye (ACE) were imaged with FOCI, in which the optical contrast of FOCI is based on intrinsic variations of the index of refraction resulting in a faster tomographic acquisition. In addition, the phase sensitivity allows simultaneous label-free acquisition of vascularisation. RESULTS We demonstrate that FOCI allows longitudinal quantification of progressive autoimmune insulitis, including the three-dimensional quantification of beta cell volume, inflammation and vascularisation. The substantially increased backscattering of islets is dominated by the insulin-zinc nanocrystals in the beta cell granules. This translates into a high specificity for the functional beta cell volume of islets. Applying FOCI to a spontaneous mouse model of type 1 diabetes, we quantify the modifications of the pancreatic microvasculature accompanying the progression of diabetes and reveal a strong correlation between increasing insulitis and density of the vascular network of the islet. CONCLUSIONS/INTERPRETATION FOCI provides a novel imaging technique for investigating functional and structural diabetes-induced alterations of the islets. The label-free detection of beta cell volume and infiltration together with vascularisation offers a unique extension to study ACE-transplanted human islets. These results are contributing to a deeper understanding of human islet transplant rejection and label-free in vivo monitoring of drug efficacy.
Collapse
Affiliation(s)
- Corinne Berclaz
- Laboratoire d'Optique Biomédicale, Ecole Polytechnique Fédérale de Lausanne, CH1015, Lausanne, Switzerland
| | | | - Daniel Szlag
- Laboratoire d'Optique Biomédicale, Ecole Polytechnique Fédérale de Lausanne, CH1015, Lausanne, Switzerland
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, Torun, Poland
| | - Jerome Extermann
- Laboratoire d'Optique Biomédicale, Ecole Polytechnique Fédérale de Lausanne, CH1015, Lausanne, Switzerland
- Hepia, University of Applied Science of Western Switzerland, Genève, Switzerland
| | - Lisbeth Hansen
- EMV - Immunology, Lund University, BMC, D14, 221 84, Lund, Sweden
| | - Arno Bouwens
- Laboratoire d'Optique Biomédicale, Ecole Polytechnique Fédérale de Lausanne, CH1015, Lausanne, Switzerland
| | - Martin Villiger
- Laboratoire d'Optique Biomédicale, Ecole Polytechnique Fédérale de Lausanne, CH1015, Lausanne, Switzerland
| | - Joan Goulley
- Swiss Institute for Experimental Cancer Research, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Frans Schuit
- Gene Expression Unit, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | - Theo Lasser
- Laboratoire d'Optique Biomédicale, Ecole Polytechnique Fédérale de Lausanne, CH1015, Lausanne, Switzerland.
| | - Dan Holmberg
- EMV - Immunology, Lund University, BMC, D14, 221 84, Lund, Sweden.
| |
Collapse
|
45
|
Non-invasive cell type selective in vivo monitoring of insulin resistance dynamics. Sci Rep 2016; 6:21448. [PMID: 26899548 PMCID: PMC4761884 DOI: 10.1038/srep21448] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 01/22/2016] [Indexed: 01/18/2023] Open
Abstract
Insulin resistance contributes to the development of cardio-vascular disease and diabetes. An important but unresolved task is to study the dynamics of insulin resistance in selective cell types of insulin target tissues in vivo. Here we present a novel technique to monitor insulin resistance dynamics non-invasively and longitudinally in vivo in a cell type-specific manner, exemplified by the pancreatic β-cell situated within the micro-organ the islet of Langerhans. We utilize the anterior chamber of the eye (ACE) as a transplantation site and the cornea as a natural body-window to study the development and reversibility of insulin resistance. Engrafted islets in the ACE that express a FoxO1-GFP-based biosensor in their β-cells, report on insulin resistance measured by fluorescence microscopy at single-cell resolution in the living mouse. This technique allows monitoring of cell type specific insulin sensitivity/resistance in real-time in the context of whole body insulin resistance during progression and intervention of disease.
Collapse
|
46
|
Arrojo e Drigo R, Ali Y, Diez J, Srinivasan DK, Berggren PO, Boehm BO. New insights into the architecture of the islet of Langerhans: a focused cross-species assessment. Diabetologia 2015. [PMID: 26215305 DOI: 10.1007/s00125-015-3699-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The human genome project and its search for factors underlying human diseases has fostered a major human research effort. Therefore, unsurprisingly, in recent years we have observed an increasing number of studies on human islet cells, including disease approaches focusing on type 1 and type 2 diabetes. Yet, the field of islet and diabetes research relies on the legacy of rodent-based investigations, which have proven difficult to translate to humans, particularly in type 1 diabetes. Whole islet physiology and pathology may differ between rodents and humans, and thus a comprehensive cross-species as well as species-specific view on islet research is much needed. In this review we summarise the current knowledge of interspecies islet cytoarchitecture, and discuss its potential impact on islet function and future perspectives in islet pathophysiology research.
Collapse
Affiliation(s)
- Rafael Arrojo e Drigo
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore
| | - Yusuf Ali
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore
| | - Juan Diez
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore
| | - Dinesh Kumar Srinivasan
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore
| | - Per-Olof Berggren
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore.
- Imperial College London, London, UK.
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska University Hospital L1, Karolinska Institutet, SE-171 76, Stockholm, Sweden.
| | - Bernhard O Boehm
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore.
- Imperial College London, London, UK.
- Department of Internal Medicine 1, Ulm University Medical Centre, Ulm, Germany.
| |
Collapse
|
47
|
Choi M, Kwok SJJ, Yun SH. In vivo fluorescence microscopy: lessons from observing cell behavior in their native environment. Physiology (Bethesda) 2015; 30:40-9. [PMID: 25559154 DOI: 10.1152/physiol.00019.2014] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Microscopic imaging techniques to visualize cellular behaviors in their natural environment play a pivotal role in biomedical research. Here, we review how recent technical advances in intravital microscopy have enabled unprecedented access to cellular physiology in various organs of mice in normal and diseased states.
Collapse
Affiliation(s)
- Myunghwan Choi
- Harvard Medical School and Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts; and
| | - Sheldon J J Kwok
- Harvard Medical School and Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts; and Harvard-MIT Health Sciences and Technology, Cambridge, Massachusetts
| | - Seok Hyun Yun
- Harvard Medical School and Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts; and Harvard-MIT Health Sciences and Technology, Cambridge, Massachusetts
| |
Collapse
|
48
|
Peppin JF, Albrecht PJ, Argoff C, Gustorff B, Pappagallo M, Rice FL, Wallace MS. Skin Matters: A Review of Topical Treatments for Chronic Pain. Part Two: Treatments and Applications. Pain Ther 2015; 4:33-50. [PMID: 25630651 PMCID: PMC4470969 DOI: 10.1007/s40122-015-0032-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Indexed: 12/26/2022] Open
Abstract
In Part One of this two-part series, we discussed skin physiology and anatomy as well as generalities concerning topical analgesics. This modality of therapy has lesser side effects and drug-drug interactions, and patients tolerate this form of therapy better than many oral options. Unfortunately, this modality is not used as often as it could be in chronic pain states, such as that from neuropathic pain. Part Two discusses specific therapies, local anesthetics, and other drugs, as well as how a clinician might use specific aspects of a patient's neuropathic pain presentation to help guide them in the selection of a topical agent.
Collapse
Affiliation(s)
- John F Peppin
- Center for Bioethics Pain Management and Medicine, St. Louis, MO, USA,
| | | | | | | | | | | | | |
Collapse
|
49
|
Nacher M, Estil Les E, Garcia A, Nadal B, Pairó M, Garcia C, Secanella L, Novials A, Montanya E. Human Serum Versus Human Serum Albumin Supplementation in Human Islet Pretransplantation Culture: In Vitro and In Vivo Assessment. Cell Transplant 2015; 25:343-52. [PMID: 25955150 DOI: 10.3727/096368915x688119] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
There is conflicting evidence favoring both the use of human serum (HS) and of human serum albumin (HSA) in human islet culture. We evaluated the effects of HS versus HSA supplementation on 1) in vitro β-cell viability and function and 2) in vivo islet graft revascularization, islet viability, β-cell death, and metabolic outcome after transplantation. Islets isolated from 14 cadaveric organ donors were cultured for 3 days in CMRL 1066 medium supplemented with HS or HSA. After 3 days in culture, β-cell apoptosis was lower in HS group (1.41 ± 0.27 vs. 2.38 ± 0.39%, p = 0.029), and the recovery of islets was 77 ± 11% and 54 ± 1% in HS- and HSA-cultured groups, respectively. Glucose-stimulated insulin secretion (GSIS) was higher in HS group (29.4, range 10.4-99.9, vs. 22.3, range 8.7-70.6, p = 0.031). In vivo viability and revascularization was determined in HS- and HSA-cultured islets transplanted into the anterior chamber of the eye of Balb/c mice (n = 14), and β-cell apoptosis in paraffin-embedded mouse eyes. Islet viability and β-cell apoptosis were similar in both groups. Revascularization was observed in one graft (HS group) on day 10 after transplantation. Islet function was determined in streptozotocin (STZ)-diabetic nude mice (n = 33) transplanted with 2,000 IEQs cultured with HS or HSA that showed similar blood glucose levels and percentage of normoglycemic animals over time. In conclusion, human islets cultured in medium supplemented with HS showed higher survival in vitro, as well as islet viability and function. The higher in vitro survival increased the number of islets available for transplantation. However, the beneficial effect on viability and function did not translate into an improved metabolic evolution when a similar number of HSA- and HS-cultured islets was transplanted.
Collapse
Affiliation(s)
- Montserrat Nacher
- Hospital Universitari Bellvitge-IDIBELLL, Hospitalet de Llobregat, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Guldner IH, Zhang S. A journey to uncharted territory: new technical frontiers in studying tumor-stromal cell interactions. Integr Biol (Camb) 2015; 7:153-61. [PMID: 25500646 PMCID: PMC4324098 DOI: 10.1039/c4ib00192c] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The crosstalk between tumor cells and cells of the tumor stroma dictate malignant progression and represent an intriguing and viable anticancer therapeutic target. The successful development of therapeutics targeting tumor-stroma interactions is tied to the insight provided by basic research on such crosstalk. Tumor-stroma interactions can be transient and dynamic, and they occur within defined spatiotemporal contexts among genetically and compositionally heterogeneous populations of cells, yet methods currently applied to study the said crosstalk do not sufficiently address these features. Emerging imaging and genetic methods, however, can overcome limitations of traditional approaches and provide unprecedented insight into tumor-stroma crosstalk with unparalleled accuracy. The comprehensive data obtained by applying emerging methods will require processing and analysis by multidisciplinary teams, but the efforts will ultimately rejuvenate hope in developing novel therapies against pro-tumorigenic tumor-stroma crosstalk.
Collapse
Affiliation(s)
- Ian H Guldner
- Department of Biological Science, Harper Cancer Research Institute, University of Notre Dame, A130 Harper Hall, Notre Dame, IN 46556, USA.
| | | |
Collapse
|