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
|
Mateus Gonçalves L, Andrade Barboza C, Almaça J. Diabetes as a Pancreatic Microvascular Disease-A Pericytic Perspective. J Histochem Cytochem 2024; 72:131-148. [PMID: 38454609 PMCID: PMC10956440 DOI: 10.1369/00221554241236535] [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: 11/28/2023] [Accepted: 02/09/2024] [Indexed: 03/09/2024] Open
Abstract
Diabetes is not only an endocrine but also a vascular disease. Vascular defects are usually seen as consequence of diabetes. However, at the level of the pancreatic islet, vascular alterations have been described before symptom onset. Importantly, the cellular and molecular mechanisms underlying these early vascular defects have not been identified, neither how these could impact the function of islet endocrine cells. In this review, we will discuss the possibility that dysfunction of the mural cells of the microvasculature-known as pericytes-underlies vascular defects observed in islets in pre-symptomatic stages. Pericytes are crucial for vascular homeostasis throughout the body, but their physiological and pathophysiological functions in islets have only recently started to be explored. A previous study had already raised interest in the "microvascular" approach to this disease. With our increased understanding of the crucial role of the islet microvasculature for glucose homeostasis, here we will revisit the vascular aspects of islet function and how their deregulation could contribute to diabetes pathogenesis, focusing in particular on type 1 diabetes (T1D).
Collapse
Affiliation(s)
- Luciana Mateus Gonçalves
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Catarina Andrade Barboza
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Joana Almaça
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida
- Molecular and Cellular Pharmacology Graduate Program, University of Miami Miller School of Medicine, Miami, Florida
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
5
|
James EA, Joglekar AV, Linnemann AK, Russ HA, Kent SC. The beta cell-immune cell interface in type 1 diabetes (T1D). Mol Metab 2023; 78:101809. [PMID: 37734713 PMCID: PMC10622886 DOI: 10.1016/j.molmet.2023.101809] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/01/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND T1D is an autoimmune disease in which pancreatic islets of Langerhans are infiltrated by immune cells resulting in the specific destruction of insulin-producing islet beta cells. Our understanding of the factors leading to islet infiltration and the interplay of the immune cells with target beta cells is incomplete, especially in human disease. While murine models of T1D have provided crucial information for both beta cell and autoimmune cell function, the translation of successful therapies in the murine model to human disease has been a challenge. SCOPE OF REVIEW Here, we discuss current state of the art and consider knowledge gaps concerning the interface of the islet beta cell with immune infiltrates, with a focus on T cells. We discuss pancreatic and immune cell phenotypes and their impact on cell function in health and disease, which we deem important to investigate further to attain a more comprehensive understanding of human T1D disease etiology. MAJOR CONCLUSIONS The last years have seen accelerated development of approaches that allow comprehensive study of human T1D. Critically, recent studies have contributed to our revised understanding that the pancreatic beta cell assumes an active role, rather than a passive position, during autoimmune disease progression. The T cell-beta cell interface is a critical axis that dictates beta cell fate and shapes autoimmune responses. This includes the state of the beta cell after processing internal and external cues (e.g., stress, inflammation, genetic risk) that that contributes to the breaking of tolerance by hyperexpression of human leukocyte antigen (HLA) class I with presentation of native and neoepitopes and secretion of chemotactic factors to attract immune cells. We anticipate that emerging insights about the molecular and cellular aspects of disease initiation and progression processes will catalyze the development of novel and innovative intervention points to provide additional therapies to individuals affected by T1D.
Collapse
Affiliation(s)
- Eddie A James
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Alok V Joglekar
- Center for Systems Immunology and Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amelia K Linnemann
- Center for Diabetes and Metabolic Diseases, and Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Holger A Russ
- Diabetes Institute, University of Florida, Gainesville, FL, USA; Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Sally C Kent
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
6
|
Thompson PJ, Pipella J, Rutter GA, Gaisano HY, Santamaria P. Islet autoimmunity in human type 1 diabetes: initiation and progression from the perspective of the beta cell. Diabetologia 2023; 66:1971-1982. [PMID: 37488322 PMCID: PMC10542715 DOI: 10.1007/s00125-023-05970-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/30/2023] [Indexed: 07/26/2023]
Abstract
Type 1 diabetes results from the poorly understood process of islet autoimmunity, which ultimately leads to the loss of functional pancreatic beta cells. Mounting evidence supports the notion that the activation and evolution of islet autoimmunity in genetically susceptible people is contingent upon early life exposures affecting the islets, especially beta cells. Here, we review some of the recent advances and studies that highlight the roles of these changes as well as antigen presentation and stress response pathways in beta cells in the onset and propagation of the autoimmune process in type 1 diabetes. Future progress in this area holds promise for advancing islet- and beta cell-directed therapies that could be implemented in the early stages of the disease and could be combined with immunotherapies.
Collapse
Affiliation(s)
- Peter J Thompson
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB, Canada.
| | - Jasmine Pipella
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| | - Guy A Rutter
- CRCHUM and Department of Medicine, Université de Montréal, Montréal, QC, Canada.
- Department of Diabetes, Endocrinology and Medicine, Faculty of Medicine, Imperial College, London, UK.
- LKC School of Medicine, Nanyang Technological College, Singapore, Republic of Singapore.
| | - Herbert Y Gaisano
- Departments of Medicine and Physiology, University of Toronto, Toronto, ON, Canada
| | - Pere Santamaria
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
7
|
Mateus Gonçalves L, Fahd Qadir MM, Boulina M, Makhmutova M, Pereira E, Almaça J. Pericyte dysfunction and impaired vasomotion are hallmarks of islets during the pathogenesis of type 1 diabetes. Cell Rep 2023; 42:112913. [PMID: 37531253 PMCID: PMC10529889 DOI: 10.1016/j.celrep.2023.112913] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/26/2023] [Accepted: 07/16/2023] [Indexed: 08/04/2023] Open
Abstract
Pancreatic islets are endocrine organs that depend on their microvasculature to function. Along with endothelial cells, pericytes comprise the islet microvascular network. These mural cells are crucial for microvascular stability and function, but it is not known if/how they are affected during the development of type 1 diabetes (T1D). Here, we investigate islet pericyte density, phenotype, and function using living pancreas slices from donors without diabetes, donors with a single T1D-associated autoantibody (GADA+), and recent onset T1D cases. Our data show that islet pericyte and capillary responses to vasoactive stimuli are impaired early on in T1D. Microvascular dysfunction is associated with a switch in the phenotype of islet pericytes toward myofibroblasts. Using publicly available RNA sequencing (RNA-seq) data, we further found that transcriptional alterations related to endothelin-1 signaling and vascular and extracellular matrix (ECM) remodeling are hallmarks of single autoantibody (Aab)+ donor pancreata. Our data show that microvascular dysfunction is present at early stages of islet autoimmunity.
Collapse
Affiliation(s)
- Luciana Mateus Gonçalves
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mirza Muhammad Fahd Qadir
- Section of Endocrinology and Metabolism, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA; Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA; Tulane Center of Excellence in Sex-Based Biology & Medicine, New Orleans, LA, USA
| | - Maria Boulina
- Diabetes Research Institute, University of Miami, Miami, FL, USA
| | - Madina Makhmutova
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Elizabeth Pereira
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Joana Almaça
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA; Molecular and Cellular Pharmacology Graduate Program, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
8
|
Wang Z, Archang M, Gurlo T, Wong E, Fraser SE, Butler PC. Application of fluorescence lifetime imaging microscopy to monitor glucose metabolism in pancreatic islets in vivo. BIOMEDICAL OPTICS EXPRESS 2023; 14:4170-4178. [PMID: 37799700 PMCID: PMC10549748 DOI: 10.1364/boe.493722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/09/2023] [Accepted: 07/10/2023] [Indexed: 10/07/2023]
Abstract
Glucose stimulated insulin secretion is mediated by glucose metabolism via oxidative phosphorylation generating ATP that triggers membrane depolarization and exocytosis of insulin. In stressed beta cells, glucose metabolism is remodeled, with enhanced glycolysis uncoupled from oxidative phosphorylation, resulting in the impaired glucose-mediated insulin secretion characteristic of diabetes. Relative changes in glycolysis and oxidative phosphorylation can be monitored in living cells using the 3-component fitting approach of fluorescence lifetime imaging microscopy (FLIM). We engrafted pancreatic islets onto the iris to permit in vivo FLIM monitoring of the trajectory of glucose metabolism. The results show increased oxidative phosphorylation of islet cells (∼90% beta cells) in response to hyperglycemia; in contrast red blood cells traversing the islets maintained exclusive glycolysis as expected in the absence of mitochondria.
Collapse
Affiliation(s)
- Zhongying Wang
- Larry L. Hillblom Islet Research Center,
University of California Los Angeles, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Maani Archang
- Larry L. Hillblom Islet Research Center,
University of California Los Angeles, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Tatyana Gurlo
- Larry L. Hillblom Islet Research Center,
University of California Los Angeles, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Elaine Wong
- Larry L. Hillblom Islet Research Center,
University of California Los Angeles, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Scott E. Fraser
- Department of Biological Sciences, Bridge Institute, David Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Peter C. Butler
- Larry L. Hillblom Islet Research Center,
University of California Los Angeles, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
9
|
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
|
10
|
Uncommon Transplantation Sites: Transplantation of Islets and Islet Organoids in the Anterior Chamber of the Eye of Rodents and Monkeys. Methods Mol Biol 2022; 2592:21-36. [PMID: 36507983 DOI: 10.1007/978-1-0716-2807-2_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The anterior chamber of the eye is a highly vascularized and innervated location that is also particularly rich in oxygen and immune privileged. This uncommon transplantation site offers unique possibilities for the observation of the transplanted material as well as for local pharmacological intervention. Transplantation of islets and islet organoids to the anterior chamber of the eye of mice and monkeys facilitates a multitude of new approaches for research into islet physiology and pathophysiology and for the treatment of diabetes. We now present a short overview of the experimental possibilities and describe an updated protocol for transplantation of islets and islet organoids into mice and monkeys.
Collapse
|
11
|
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
|
12
|
Shi Y, Zhao YZ, Jiang Z, Wang Z, Wang Q, Kou L, Yao Q. Immune-Protective Formulations and Process Strategies for Improved Survival and Function of Transplanted Islets. Front Immunol 2022; 13:923241. [PMID: 35903090 PMCID: PMC9315421 DOI: 10.3389/fimmu.2022.923241] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/13/2022] [Indexed: 11/25/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease caused by the immune system attacking and destroying insulin-producing β cells in the pancreas. Islet transplantation is becoming one of the most promising therapies for T1D patients. However, its clinical use is limited by substantial cell loss after islet infusion, closely related to immune reactions, including instant blood-mediated inflammatory responses, oxidative stress, and direct autoimmune attack. Especially the grafted islets are not only exposed to allogeneic immune rejection after transplantation but are also subjected to an autoimmune process that caused the original disease. Due to the development and convergence of expertise in biomaterials, nanotechnology, and immunology, protective strategies are being investigated to address this issue, including exploring novel immune protective agents, encapsulating islets with biomaterials, and searching for alternative implantation sites, or co-transplantation with functional cells. These methods have significantly increased the survival rate and function of the transplanted islets. However, most studies are still limited to animal experiments and need further studies. In this review, we introduced the immunological challenges for islet graft and summarized the recent developments in immune-protective strategies to improve the outcomes of islet transplantation.
Collapse
Affiliation(s)
- Yannan Shi
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ying-Zheng Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zhikai Jiang
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zeqing Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qian Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Longfa Kou
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Qing Yao, ; Longfa Kou,
| | - Qing Yao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Qing Yao, ; Longfa Kou,
| |
Collapse
|
13
|
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
|
14
|
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
|
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
|
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
|
20
|
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
|
21
|
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
|
22
|
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
|