1
|
Pignatelli C, Campo F, Neroni A, Piemonti L, Citro A. Bioengineering the Vascularized Endocrine Pancreas: A Fine-Tuned Interplay Between Vascularization, Extracellular-Matrix-Based Scaffold Architecture, and Insulin-Producing Cells. Transpl Int 2022; 35:10555. [PMID: 36090775 PMCID: PMC9452644 DOI: 10.3389/ti.2022.10555] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022]
Abstract
Intrahepatic islet transplantation is a promising β-cell replacement strategy for the treatment of type 1 diabetes. Instant blood-mediated inflammatory reactions, acute inflammatory storm, and graft revascularization delay limit islet engraftment in the peri-transplant phase, hampering the success rate of the procedure. Growing evidence has demonstrated that islet engraftment efficiency may take advantage of several bioengineering approaches aimed to recreate both vascular and endocrine compartments either ex vivo or in vivo. To this end, endocrine pancreas bioengineering is an emerging field in β-cell replacement, which might provide endocrine cells with all the building blocks (vascularization, ECM composition, or micro/macro-architecture) useful for their successful engraftment and function in vivo. Studies on reshaping either the endocrine cellular composition or the islet microenvironment have been largely performed, focusing on a single building block element, without, however, grasping that their synergistic effect is indispensable for correct endocrine function. Herein, the review focuses on the minimum building blocks that an ideal vascularized endocrine scaffold should have to resemble the endocrine niche architecture, composition, and function to foster functional connections between the vascular and endocrine compartments. Additionally, this review highlights the possibility of designing bioengineered scaffolds integrating alternative endocrine sources to overcome donor organ shortages and the possibility of combining novel immune-preserving strategies for long-term graft function.
Collapse
Affiliation(s)
- Cataldo Pignatelli
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Campo
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Alessia Neroni
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Lorenzo Piemonti
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Antonio Citro
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
2
|
Anti-Inflammatory Strategies in Intrahepatic Islet Transplantation: A Comparative Study in Preclinical Models. Transplantation 2018; 102:240-248. [PMID: 28902069 DOI: 10.1097/tp.0000000000001925] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND The identification of pathway(s) playing a pivotal role in peritransplant detrimental inflammatory events represents the crucial step toward a better management and outcome of pancreatic islet transplanted patients. Recently, we selected the CXCR1/2 inhibition as a relevant strategy in enhancing pancreatic islet survival after transplantation. METHODS Here, the most clinically used anti-inflammatory compounds (IL1-receptor antagonist, steroids, and TNF-α inhibitor) alone or in combination with a CXCR1/2 inhibitor were evaluated in their ability to improve engraftment or delay graft rejection. To rule out bias related to transplantation site, we used well-established preclinical syngeneic (250 C57BL/6 equivalent islets in C57BL/6) and allogeneic (400 Balb/c equivalent islets in C57BL6) intrahepatic islet transplantation platforms. RESULTS In mice, we confirmed that targeting the CXCR1/2 pathway is crucial in preserving islet function and improving engraftment. In the allogeneic setting, CXCR1/2 inhibitor alone could reduce the overall recruitment of transplant-induced leukocytes and significantly prolong the time to graft rejection both as a single agent and in combination with immunosuppression. No other anti-inflammatory compounds tested (IL1-receptor antagonist, steroids, and TNF-α inhibitor) alone or in combination with CXCR1/2 inhibitor improve islet engraftment and significantly delay graft rejection in the presence of MMF + FK-506 immunosuppressive treatment. CONCLUSIONS These findings indicate that only the CXCR1/2-mediated axis plays a crucial role in controlling the islet damage and should be a target for intervention to improve the efficiency of islet transplantation.
Collapse
|
3
|
Ribback S, Sonke J, Lohr A, Frohme J, Peters K, Holm J, Peters M, Cigliano A, Calvisi DF, Dombrowski F. Hepatocellular glycogenotic foci after combined intraportal pancreatic islet transplantation and knockout of the carbohydrate responsive element binding protein in diabetic mice. Oncotarget 2017; 8:104315-104329. [PMID: 29262643 PMCID: PMC5732809 DOI: 10.18632/oncotarget.22234] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/05/2017] [Indexed: 01/01/2023] Open
Abstract
Aims The intraportal pancreatic islet transplantation (IPIT) model of diabetic rats is an insulin mediated model of hepatocarcinogenesis characterized by the induction of clear cell foci (CCF) of altered hepatocytes, which are pre-neoplastic lesions excessively storing glycogen (glycogenosis) and exhibiting activation of the AKT/mTOR protooncogenic pathway. In this study, we transferred the IPIT model to the mouse and combined it with the knockout of the transcription factor carbohydrate responsive element binding protein (chREBP). Methods C57BL/6J Wild-type (WT) and chREBP-knockout (chREBP-KO) mice (n = 297) were matched to 16 groups (WT/ chREBP-KO, experimental/control, streptozotocine-induced diabetic/not diabetic, one/four weeks). Experimental groups received the intraportal transplantation of 70 pancreatic islets. Liver and pancreatic tissue was examined using histology, morphometry, enzyme- and immunohistochemistry and electron microscopy. Results CCF emerged in the liver acini downstream of the transplanted islets. In comparison to WT lesions, CCF of chREBP-KO mice displayed more glycogen accumulation, reduced activity of the gluconeogenic enzyme glucose-6-phosphatase, decreased glycolysis, lipogenesis and reduced levels of the AKT/mTOR cascade members. Proliferative activity of CCF was ∼two folds higher in WT mice than in chREBP-KO mice. Conclusions The IPIT model is applicable to mice, as murine CCF resemble preneoplastic liver lesions from this hepatocarcinogenesis model in the rat in terms of morphological, metabolic and molecular alterations and proliferative activity, which is diminished after chREBP knockout. chREBP appears to be an essential component of AKT/mTOR mediated cell proliferation and the metabolic switch from a glycogenotic to lipogenic phenotype in precursor lesions of hepatocarcinogenesis.
Collapse
Affiliation(s)
- Silvia Ribback
- Institut für Pathologie, Universitaetsmedizin Greifswald, Greifswald, Germany
| | - Jenny Sonke
- Institut für Pathologie, Universitaetsmedizin Greifswald, Greifswald, Germany
| | - Andrea Lohr
- Institut für Pathologie, Universitaetsmedizin Greifswald, Greifswald, Germany
| | - Josephine Frohme
- Institut für Pathologie, Universitaetsmedizin Greifswald, Greifswald, Germany
| | - Kristin Peters
- Institut für Pathologie, Universitaetsmedizin Greifswald, Greifswald, Germany
| | - Johannes Holm
- Institut für Pathologie, Universitaetsmedizin Greifswald, Greifswald, Germany
| | - Michele Peters
- Institut für Pathologie, Universitaetsmedizin Greifswald, Greifswald, Germany
| | - Antonio Cigliano
- Institut für Pathologie, Universitaetsmedizin Greifswald, Greifswald, Germany
| | - Diego F Calvisi
- Institut für Pathologie, Universitaetsmedizin Greifswald, Greifswald, Germany
| | - Frank Dombrowski
- Institut für Pathologie, Universitaetsmedizin Greifswald, Greifswald, Germany
| |
Collapse
|
4
|
Costa OR, Stangé G, Verhaeghen K, Brackeva B, Nonneman E, Hampe CS, Ling Z, Pipeleers D, Gorus FK, Martens GA. Development of an Enhanced Sensitivity Bead-Based Immunoassay for Real-Time In Vivo Detection of Pancreatic β-Cell Death. Endocrinology 2015; 156:4755-60. [PMID: 26431226 PMCID: PMC5393343 DOI: 10.1210/en.2015-1636] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
There is a clinical need for plasma tests to detect and quantify the in vivo destruction of pancreatic β-cells in type 1 diabetes. We previously developed a time-resolved fluorescence immunoassay (TRFIA) to glutamate decarboxylase 65 kDa (GAD65) (GAD65-TRFIA) that was able to detect the synchronous necrotic destruction of transplanted β-cells in the hours after their infusion in the liver. This GAD65-TRFIA, however, lacked sensitivity to detect continued β-cell rejection beyond this acute phase. The aim of present study was to gain at least an order of magnitude in analytical sensitivity by switching to Becton Dickinson cytometric bead array (CBA) (GAD65-CBA) enhanced sensitivity format, using the same couple of monoclonal antibodies. We compared the performances of GAD65-CBA and GAD65-TRFIA using Clinical and Laboratory Standards Institute protocols for linearity, imprecision, specificity, limit of detection, and functional sensitivity. We conducted a method comparison and assessed the biologic potential on samples from human recipients of islet grafts. The GAD65-CBA showed acceptable linearity and imprecision. Switching from TRFIA to CBA lowered functional sensitivity by a factor 35 and lowered limit of detection by a factor 11 with minimal need for method optimization. The enhanced sensitivity greatly expands the application domain of our biomarker and allowed for the first time to detect ongoing β-cell destruction up to at least 1 day after islet transplantation. We conclude that the GAD65-CBA is suitable for biological and clinical assessment of the real-time destruction of β-cells in intraportal transplantation.
Collapse
Affiliation(s)
- Olivier R Costa
- Diabetes Research Center (O.C., G.S., B.B., E.N., Z.L., D.P., F.K.G., G.A.M.), Brussels Free University and Department of Clinical Chemistry and Radio-Immunology (O.C., K.V., B.B., F.K.G., G.A.M.), Universitair Ziekenhuis Brussel, B1090 Brussels, Belgium; and Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109-4725
| | - Geert Stangé
- Diabetes Research Center (O.C., G.S., B.B., E.N., Z.L., D.P., F.K.G., G.A.M.), Brussels Free University and Department of Clinical Chemistry and Radio-Immunology (O.C., K.V., B.B., F.K.G., G.A.M.), Universitair Ziekenhuis Brussel, B1090 Brussels, Belgium; and Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109-4725
| | - Katrijn Verhaeghen
- Diabetes Research Center (O.C., G.S., B.B., E.N., Z.L., D.P., F.K.G., G.A.M.), Brussels Free University and Department of Clinical Chemistry and Radio-Immunology (O.C., K.V., B.B., F.K.G., G.A.M.), Universitair Ziekenhuis Brussel, B1090 Brussels, Belgium; and Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109-4725
| | - Benedicte Brackeva
- Diabetes Research Center (O.C., G.S., B.B., E.N., Z.L., D.P., F.K.G., G.A.M.), Brussels Free University and Department of Clinical Chemistry and Radio-Immunology (O.C., K.V., B.B., F.K.G., G.A.M.), Universitair Ziekenhuis Brussel, B1090 Brussels, Belgium; and Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109-4725
| | - Ellen Nonneman
- Diabetes Research Center (O.C., G.S., B.B., E.N., Z.L., D.P., F.K.G., G.A.M.), Brussels Free University and Department of Clinical Chemistry and Radio-Immunology (O.C., K.V., B.B., F.K.G., G.A.M.), Universitair Ziekenhuis Brussel, B1090 Brussels, Belgium; and Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109-4725
| | - Christiane S Hampe
- Diabetes Research Center (O.C., G.S., B.B., E.N., Z.L., D.P., F.K.G., G.A.M.), Brussels Free University and Department of Clinical Chemistry and Radio-Immunology (O.C., K.V., B.B., F.K.G., G.A.M.), Universitair Ziekenhuis Brussel, B1090 Brussels, Belgium; and Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109-4725
| | - Zhidong Ling
- Diabetes Research Center (O.C., G.S., B.B., E.N., Z.L., D.P., F.K.G., G.A.M.), Brussels Free University and Department of Clinical Chemistry and Radio-Immunology (O.C., K.V., B.B., F.K.G., G.A.M.), Universitair Ziekenhuis Brussel, B1090 Brussels, Belgium; and Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109-4725
| | - Daniel Pipeleers
- Diabetes Research Center (O.C., G.S., B.B., E.N., Z.L., D.P., F.K.G., G.A.M.), Brussels Free University and Department of Clinical Chemistry and Radio-Immunology (O.C., K.V., B.B., F.K.G., G.A.M.), Universitair Ziekenhuis Brussel, B1090 Brussels, Belgium; and Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109-4725
| | - Frans K Gorus
- Diabetes Research Center (O.C., G.S., B.B., E.N., Z.L., D.P., F.K.G., G.A.M.), Brussels Free University and Department of Clinical Chemistry and Radio-Immunology (O.C., K.V., B.B., F.K.G., G.A.M.), Universitair Ziekenhuis Brussel, B1090 Brussels, Belgium; and Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109-4725
| | - Geert A Martens
- Diabetes Research Center (O.C., G.S., B.B., E.N., Z.L., D.P., F.K.G., G.A.M.), Brussels Free University and Department of Clinical Chemistry and Radio-Immunology (O.C., K.V., B.B., F.K.G., G.A.M.), Universitair Ziekenhuis Brussel, B1090 Brussels, Belgium; and Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109-4725
| |
Collapse
|
5
|
Choong FJ, Freeman C, Parish CR, Simeonovic CJ. Islet heparan sulfate but not heparan sulfate proteoglycan core protein is lost during islet isolation and undergoes recovery post-islet transplantation. Am J Transplant 2015; 15:2851-64. [PMID: 26104150 DOI: 10.1111/ajt.13366] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 03/29/2015] [Accepted: 04/19/2015] [Indexed: 01/25/2023]
Abstract
Islet beta cells in situ express intracellular heparan sulfate (HS), a property previously shown in vitro to be important for their survival. We report that HS levels inside islet beta cells correlate with the novel intracellular localization of the HSPG core proteins for collagen type XVIII (Col18), a conventional extracellular matrix component. Syndecan-1 (Sdc1) and CD44 core proteins were similarly localized inside beta cells. During isolation, mouse islets selectively lose HS to 11-27% of normal levels but retain their HSPG core proteins. Intra-islet HS failed to recover substantially during culture for 4 days and was not reconstituted in vitro using HS mimetics. In contrast, significant recovery of intra-islet HS to ∼40-50% of normal levels occurred by 5-10 days after isotransplantation. Loss of islet HS during the isolation procedure is independent of heparanase (a HS-degrading endoglycosidase) and due, in part, to oxidative damage. Treatment with antioxidants reduced islet cell death by ∼60% and increased the HS content of isolated islets by ∼twofold compared to untreated islets, preserving intra-islet HS to ∼60% of the normal HS content of islets in situ. These findings suggest that the preservation of islet HS during the islet isolation process may optimize islet survival posttransplant.
Collapse
Affiliation(s)
- F J Choong
- Department of Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT, Australia
| | - C Freeman
- Department of Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT, Australia
| | - C R Parish
- Department of Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT, Australia
| | - C J Simeonovic
- Department of Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT, Australia
| |
Collapse
|
6
|
Zhou J, Hu W, Tang L. Non-invasive Characterization of Immune Responses to Biomedical Implants. Ann Biomed Eng 2015; 44:693-704. [DOI: 10.1007/s10439-015-1470-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/22/2015] [Indexed: 01/08/2023]
|
7
|
Ling Z, De Pauw P, Jacobs-Tulleneers-Thevissen D, Mao R, Gillard P, Hampe CS, Martens GA, In't Veld P, Lernmark Å, Keymeulen B, Gorus F, Pipeleers D. Plasma GAD65, a Marker for Early β-Cell Loss After Intraportal Islet Cell Transplantation in Diabetic Patients. J Clin Endocrinol Metab 2015; 100:2314-21. [PMID: 25816051 PMCID: PMC5393519 DOI: 10.1210/jc.2015-1216] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CONTEXT AND OBJECTIVE Intraportal islet transplantation can restore insulin production in type 1 diabetes patients, but its effect is subject to several interfering processes. To assess the influence of β-cell loss before and during engraftment, we searched for a real-time marker of β-cell destruction. Previous studies showed that 65-kDa isoform of glutamate decarboxylase (GAD65) is discharged by chemically damaged rat β-cells. We therefore examined the utility of the GAD65 assay to detect and quantify destruction of human β-cells in vitro and in vivo. DESIGN AND PARTICIPANTS A time-resolved fluorescence immunoassay was used to measure GAD65 discharge from β-cells after administration of toxins or after intraportal transplantation. The study in patients involved type 1 diabetes recipients of 56 implants. RESULTS GAD65 was discharged from cultured human β-cells between 4 and 24 hours after acute insult and proportional to the number of dying cells. It was also detected in plasma during the first 24 hours after intraportal transplantation of human islet cell grafts. Diabetic nude rat recipients without hyperglycemic correction exhibited higher plasma GAD65 levels than those with normalization. In type 1 diabetes recipients of grafts with 2-5 × 10(6) β-cells per kilogram of body weight, five of six with plasma GAD65 greater than 1 ng/mL failed to increase plasma C-peptide by greater than 0.5 ng/mL at posttransplant month 2, whereas five of six with undetectable plasma GAD 65 and 15 of 19 with intermediate levels did result in such increase. CONCLUSION Plasma GAD65 qualifies as a marker for early β-cell loss after intraportal transplantation. Further studies are needed to extend its clinical utility.
Collapse
Affiliation(s)
- Zhidong Ling
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Pieter De Pauw
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Daniel Jacobs-Tulleneers-Thevissen
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Rui Mao
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Pieter Gillard
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Christiane S Hampe
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Geert A Martens
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Peter In't Veld
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Åke Lernmark
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Bart Keymeulen
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Frans Gorus
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Daniel Pipeleers
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| |
Collapse
|
8
|
Brackeva B, De Punt V, Kramer G, Costa O, Verhaeghen K, Stangé G, Sadones J, Xavier C, Aerts JMFG, Gorus FK, Martens GA. Potential of UCHL1 as biomarker for destruction of pancreatic beta cells. J Proteomics 2015; 117:156-67. [PMID: 25638021 DOI: 10.1016/j.jprot.2015.01.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 12/09/2014] [Accepted: 01/09/2015] [Indexed: 01/08/2023]
Abstract
UNLABELLED There is a clinical need for plasma tests for real-time detection of beta cell destruction, as surrogate endpoint in islet transplantation and immunoprevention trials in type 1 diabetes. This study reports on the use of label-free LC-MS/MS proteomics for bottom-up selection of candidate biomarkers. Ubiquitin COOH-terminal hydrolase 1 (UCHL1) was identified as abundant protein in rat and human beta cells, showing promising beta cell-selectivity, and was selected for further validation in standardized toxicity models. In vitro, H2O2-induced necrosis of INS-1 cells and human islets resulted in intracellular UCHL1 depletion and its extracellular discharge. In vivo, streptozotocin progressively depleted UCHL1 from islet cores and in 50% of animals, an associated plasma UCHL1 surge was detected preceding the GAD65 peak. UCHL1 was cleared with a half-life of 20min. Whole-body dynamic planar imaging of (99m)-Technetium-labeled UCHL1 indicated a rapid UCHL1 uptake in the liver and spleen, followed by urinary excretion of mainly proteolytic UCHL1 fragments. We conclude that LC-MS/MS proteomics is a useful tool to prioritize biomarkers for beta cell injury with promising molar abundance. Despite its consistent UCHL1 discharge by damaged beta cells in vitro, its in vivo use might be restrained by its rapid elimination from plasma. BIOLOGICAL SIGNIFICANCE Our bottom-up LC-MS/MS proteomics represents a pragmatic approach to identify protein-type biomarkers of pancreatic beta cell injury. UCHL1 successfully passed sequential validation steps of beta cell-selectivity, antigenicity and toxic discharge in vitro. Whole-body dynamic planar imaging of radiolabeled recombinant UCHL1 indicated rapid clearance through the liver, spleen and urinary excretion of proteolytic fragments, likely explaining non-consistent detection in vivo. Integration of kinetic biomarker clearance studies in the a priori selection criteria is recommended before engaging in resource-intensive custom development of sensitive immunoassays for clinical translation.
Collapse
Affiliation(s)
- B Brackeva
- Diabetes Research Center, Vrije Universiteit Brussel (VUB), Belgium; Department of Clinical Chemistry and Radio-immunology, Universitair Ziekenhuis Brussel, Belgium
| | - V De Punt
- Diabetes Research Center, Vrije Universiteit Brussel (VUB), Belgium; Department of Clinical Chemistry and Radio-immunology, Universitair Ziekenhuis Brussel, Belgium
| | - G Kramer
- Department of Medical Biochemistry, Academisch Medisch Centrum, Amsterdam, Netherlands
| | - O Costa
- Department of Clinical Chemistry and Radio-immunology, Universitair Ziekenhuis Brussel, Belgium
| | - K Verhaeghen
- Department of Clinical Chemistry and Radio-immunology, Universitair Ziekenhuis Brussel, Belgium
| | - G Stangé
- Diabetes Research Center, Vrije Universiteit Brussel (VUB), Belgium
| | - J Sadones
- Department of Anatomopathology, Universitair Ziekenhuis Brussel, Belgium
| | - C Xavier
- In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel (VUB), Belgium
| | - J M F G Aerts
- Department of Medical Biochemistry, Academisch Medisch Centrum, Amsterdam, Netherlands
| | - F K Gorus
- Department of Clinical Chemistry and Radio-immunology, Universitair Ziekenhuis Brussel, Belgium
| | - G A Martens
- Diabetes Research Center, Vrije Universiteit Brussel (VUB), Belgium; Department of Clinical Chemistry and Radio-immunology, Universitair Ziekenhuis Brussel, Belgium.
| |
Collapse
|
9
|
Nano R, Racanicchi L, Melzi R, Mercalli A, Maffi P, Sordi V, Ling Z, Scavini M, Korsgren O, Celona B, Secchi A, Piemonti L. Human Pancreatic Islet Preparations Release HMGB1: (Ir)Relevance for Graft Engraftment. Cell Transplant 2013; 22:2175-86. [DOI: 10.3727/096368912x657783] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
High levels of donor-derived high-mobility group box 1 (HMGB1) protein have been associated with poor islet graft outcome in mouse models. The aim of our work was to determine whether HMGB1 released by human islets had independent proinflammatory effects that influence engraftment in humans. Human islet preparations contained and released HMGB1 in different amounts, as determined by Western blot and ELISA (median 17 pg/ml/IEQ/24 h; min–max 0–211, n = 74). HMGB1 release directly correlated with brain death, donor hyperamilasemia, and factors related to the pancreas digestion procedure (collagenase and digestion time). HMGB1 release was significantly positively associated with the release of other cytokines/chemokines, particularly with the highly released “proinflammatory” CXCL8/IL-8, CXCL1/GRO-α, and the IFN-γ-inducible chemokines CXCL10/IP-10 and CXCL9/MIG. HMGB1 release was not modulated by Toll-like receptor 2, 3, 4, 5, and 9 agonists or by exposure to IL-1β. When evaluated after islet transplantation, pretransplant HMGB1 release was weakly associated with the activation of the coagulation cascade (evaluated as serum cross-linked fibrin products), but not with the immediate posttransplant inflammatory response. Concordantly, HMGB1 did not affect short-term human islet function. Our data show that human islet HMGB1 release is a sign of “damaged” islets, although without any independent direct role in graft failure.
Collapse
Affiliation(s)
- Rita Nano
- San Raffaele Diabetes Research Institute (HSR-DRI), San Raffaele Scientific Institute, Milan, Italy
| | - Leda Racanicchi
- San Raffaele Diabetes Research Institute (HSR-DRI), San Raffaele Scientific Institute, Milan, Italy
| | - Raffaella Melzi
- San Raffaele Diabetes Research Institute (HSR-DRI), San Raffaele Scientific Institute, Milan, Italy
| | - Alessia Mercalli
- San Raffaele Diabetes Research Institute (HSR-DRI), San Raffaele Scientific Institute, Milan, Italy
| | - Paola Maffi
- Transplant Unit, Department of Medicine, San Raffaele Scientific Institute, Milan, Italy
| | - Valeria Sordi
- San Raffaele Diabetes Research Institute (HSR-DRI), San Raffaele Scientific Institute, Milan, Italy
| | - Zhidong Ling
- Diabetes Research Center, Brussels Free University-VUB, Brussels, Belgium
| | - Marina Scavini
- San Raffaele Diabetes Research Institute (HSR-DRI), San Raffaele Scientific Institute, Milan, Italy
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Division of Immunology, Uppsala University, Uppsala, Sweden
| | - Barbara Celona
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Antonio Secchi
- Transplant Unit, Department of Medicine, San Raffaele Scientific Institute, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Lorenzo Piemonti
- San Raffaele Diabetes Research Institute (HSR-DRI), San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
10
|
Maffi P, Balzano G, Ponzoni M, Nano R, Sordi V, Melzi R, Mercalli A, Scavini M, Esposito A, Peccatori J, Cantarelli E, Messina C, Bernardi M, Del Maschio A, Staudacher C, Doglioni C, Ciceri F, Secchi A, Piemonti L. Autologous pancreatic islet transplantation in human bone marrow. Diabetes 2013; 62:3523-31. [PMID: 23733196 PMCID: PMC3781459 DOI: 10.2337/db13-0465] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The liver is the current site of choice for pancreatic islet transplantation, even though it is far from being ideal. We recently have shown in mice that the bone marrow (BM) may be a valid alternative to the liver, and here we report a pilot study to test feasibility and safety of BM as a site for islet transplantation in humans. Four patients who developed diabetes after total pancreatectomy were candidates for the autologous transplantation of pancreatic islet. Because the patients had contraindications for intraportal infusion, islets were infused in the BM. In all recipients, islets engrafted successfully as shown by measurable posttransplantation C-peptide levels and histopathological evidence of insulin-producing cells or molecular markers of endocrine tissue in BM biopsy samples analyzed during follow-up. Thus far, we have recorded no adverse events related to the infusion procedure or the presence of islets in the BM. Islet function was sustained for the maximum follow-up of 944 days. The encouraging results of this pilot study provide new perspectives in identifying alternative sites for islet infusion in patients with type 1 diabetes. Moreover, this is the first unequivocal example of successful engraftment of endocrine tissue in the BM in humans.
Collapse
Affiliation(s)
- Paola Maffi
- Islet Transplantation Unit, Diabetes Research Institute, Ospedale San Raffaele, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Gianpaolo Balzano
- Department of Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Maurilio Ponzoni
- Department of Pathology, San Raffaele Scientific Institute, Milan, Italy
| | - Rita Nano
- Division of Immunology, Transplantation, and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
- Beta Cell Biology Unit, Diabetes Research Institute, Ospedale San Raffaele, Milan, Italy
| | - Valeria Sordi
- Division of Immunology, Transplantation, and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
- Beta Cell Biology Unit, Diabetes Research Institute, Ospedale San Raffaele, Milan, Italy
| | - Raffaella Melzi
- Division of Immunology, Transplantation, and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
- Beta Cell Biology Unit, Diabetes Research Institute, Ospedale San Raffaele, Milan, Italy
| | - Alessia Mercalli
- Division of Immunology, Transplantation, and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
- Beta Cell Biology Unit, Diabetes Research Institute, Ospedale San Raffaele, Milan, Italy
| | - Marina Scavini
- Epidemiology and Data Management Unit, Diabetes Research Institute, Ospedale San Raffaele, Milan, Italy
| | - Antonio Esposito
- Department of Radiology, San Raffaele Scientific Institute, Milan, Italy
| | - Jacopo Peccatori
- Hematology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Cantarelli
- Division of Immunology, Transplantation, and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
- Beta Cell Biology Unit, Diabetes Research Institute, Ospedale San Raffaele, Milan, Italy
| | - Carlo Messina
- Hematology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Bernardi
- Hematology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Del Maschio
- Department of Radiology, San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Carlo Staudacher
- Department of Surgery, San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Claudio Doglioni
- Department of Pathology, San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Fabio Ciceri
- Hematology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Antonio Secchi
- Vita-Salute San Raffaele University, Milan, Italy
- Clinical Transplant Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
- Corresponding author: Lorenzo Piemonti, , or Antonio Secchi,
| | - Lorenzo Piemonti
- Division of Immunology, Transplantation, and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
- Beta Cell Biology Unit, Diabetes Research Institute, Ospedale San Raffaele, Milan, Italy
- Corresponding author: Lorenzo Piemonti, , or Antonio Secchi,
| |
Collapse
|
11
|
Abstract
Early innate inflammatory reaction strongly affects islet engraftment and survival after intrahepatic transplantation. This early immune response is triggered by ischemia-reperfusion injury and instant blood mediated inflammatory reaction (IBMIR) occurring hours and days after islet infusion. Evidence in both mouse model and in human counterpart suggest the involvement of coagulation, complement system, and proinflammatory chemokines/cytokines. Identification and targeting of pathway(s), playing a role as "master regulator(s)" in post-transplant detrimental inflammatory events, is now mandatory to improve islet transplantation success. This review will focus on inflammatory pathway(s) differentially modulated by islet isolation and mainly associated with the early post-transplant events. Moreover, we will take into account anti-inflammatory strategies that have been tested at 2 levels: on the graft, ex vivo, during islet culture (i.e., donor) and/or on the graft site, in vivo, early after islet infusion (i.e., recipient).
Collapse
Affiliation(s)
- Antonio Citro
- Beta Cell Biology Unit, Diabetes Research Institute, San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy,
| | | | | |
Collapse
|
12
|
Jiang L, Brackeva B, Ling Z, Kramer G, Aerts JM, Schuit F, Keymeulen B, Pipeleers D, Gorus F, Martens GA. Potential of protein phosphatase inhibitor 1 as biomarker of pancreatic β-cell injury in vitro and in vivo. Diabetes 2013; 62:2683-8. [PMID: 23557701 PMCID: PMC3717856 DOI: 10.2337/db12-1507] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
There is a need for plasma-based tests that can directly measure the extent of β-cell injury in vivo in patients receiving islet grafts and in animal models. In this study, we propose protein phosphatase 1, regulatory (inhibitor) subunit 1A (PPP1R1A) as a novel biomarker for acute β-cell destruction. Liquid chromatography-tandem mass spectrometry proteome analysis of fluorescence-activated cell sorter-purified β-cells, tissue-comparative Western blotting, and immunohistochemistry indicated relatively high molar abundance and selectivity of PPP1R1A in β-cells. PPP1R1A was discharged into the extracellular space of chemically injured rat and human islets in vitro, proportionate to the extent of β-cell death. Streptozotocin injection in rats led to a progressive PPP1R1A depletion from the cytoplasm of disintegrating β-cells and a marked surge in plasma levels detectable by an affinity-capture method. A similar massive PPP1R1A discharge in blood was also detected in three patients immediately after intraportal islet transplantation. Our findings provide first proof-of-principle for PPP1R1A as real-time biomarker of β-cell destruction in animal models and patients and warrant development of more sensitive methods for its further validation in clinical trials.
Collapse
Affiliation(s)
- Lei Jiang
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Benedicte Brackeva
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Clinical Chemistry and Radioimmunology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Zhidong Ling
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Gertjan Kramer
- Department of Medical Biochemistry, Academisch Medisch Centrum, Amsterdam, the Netherlands
| | - Johannes M. Aerts
- Department of Medical Biochemistry, Academisch Medisch Centrum, Amsterdam, the Netherlands
| | - Frans Schuit
- Gene Expression Unit, Department of Molecular Cell Biology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Bart Keymeulen
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Daniel Pipeleers
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Frans Gorus
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Clinical Chemistry and Radioimmunology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Geert A. Martens
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Clinical Chemistry and Radioimmunology, Universitair Ziekenhuis Brussel, Brussels, Belgium
- Corresponding author: Geert A. Martens,
| |
Collapse
|
13
|
SoRelle JA, Itoh T, Peng H, Kanak MA, Sugimoto K, Matsumoto S, Levy MF, Lawrence MC, Naziruddin B. Withaferin A inhibits pro-inflammatory cytokine-induced damage to islets in culture and following transplantation. Diabetologia 2013; 56:814-24. [PMID: 23318585 DOI: 10.1007/s00125-012-2813-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 12/07/2012] [Indexed: 11/28/2022]
Abstract
AIMS/HYPOTHESIS Beta cell death triggered by pro-inflammatory cytokines plays a central role in the pathogenesis of type 1 diabetes and loss of transplanted islets. The nuclear factor κB (NF-κB) signalling pathway is a key regulator of beta cell stress response, survival and apoptosis. Withaferin A (WA), a steroidal lactone derived from Withania somnifera, has been demonstrated to be a potent, safe, anti-inflammatory molecule that can inhibit NF-κB signalling. Therefore, we evaluated the ability of WA to protect mouse and human islets from the damaging effects of pro-inflammatory cytokines in vitro and following intraportal transplantation. METHODS Mouse and human islets were treated with a cytokine cocktail, and NF-κB activation was measured by immunoblots, p65 nuclear translocation and chromatin immunoprecipitation of p65-bound DNA. Intraportal transplantation of a marginal mass of syngeneic mouse islets was performed to evaluate the in vivo protective effect of WA. RESULTS Treatment with WA substantially improved islet engraftment of syngeneic islets (83% for infusion with 200 islets + WA; 0% for 200 islets + vehicle) in a mouse model of diabetes, compared with marginal graft controls with superior islet function in WA-treated mice confirmed by glucose tolerance test. Treatment of human and mouse islets with WA prevented cytokine-induced cell death, inhibited inflammatory cytokine secretion and protected islet potency. CONCLUSIONS WA was shown to be a strong inhibitor of the inflammatory response in islets, protecting against cytokine-induced cell damage while improving survival of transplanted islets. These results suggest that WA could be incorporated as an adjunctive treatment to improve islet transplant outcome.
Collapse
Affiliation(s)
- J A SoRelle
- Institute of Biomedical Studies, Baylor University, Waco, TX, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Zhou J, Hao G, Weng H, Tsai YT, Baker DW, Sun X, Tang L. In vivo evaluation of medical device-associated inflammation using a macrophage-specific positron emission tomography (PET) imaging probe. Bioorg Med Chem Lett 2013; 23:2044-7. [PMID: 23481649 DOI: 10.1016/j.bmcl.2013.02.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 01/24/2013] [Accepted: 02/01/2013] [Indexed: 12/16/2022]
Abstract
To image implant-surrounding activated macrophages, a macrophage-specific PET probe was prepared by conjugating folic acid (FA) and 2,2',2″,2‴-(1,4,7,10-tetraazacyclododecane-1,4,7,10-tetrayl)tetracetic acid (DOTA) to polyethylene glycol (PEG) and then labeling the conjugate with Ga-68. In vivo PET imaging evaluations demonstrate that the probe is able to detect foreign body reactions, and more importantly, quantify the degree of inflammatory responses to an implanted medical device. These results were further validated by histological analysis.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
LC-MS/MS identification of doublecortin as abundant beta cell-selective protein discharged by damaged beta cells in vitro. J Proteomics 2013; 80:268-80. [PMID: 23337804 DOI: 10.1016/j.jprot.2012.12.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 12/14/2012] [Accepted: 12/26/2012] [Indexed: 02/06/2023]
Abstract
There is a clinical need for plasma tests that can directly detect injury to pancreatic beta cells in type 1 diabetes. Such tests require biomarkers that are abundantly and selectively released into plasma by damaged beta cells. We combined LC-MS/MS proteomics and tissue-comparative transcriptomics of FACS-purified beta cells for bottom-up identification of candidate markers. Less than 10% of 467 proteins detected in beta cells showed endocrine-enriched expression. One surprising candidate was the neuronal migration marker doublecortin: in situ analysis revealed uniform doublecortin expression in the cytoplasm of all beta cells. Western blotting and real-time PCR confirmed its strong beta cell-selectivity outside the brain and its high molar abundance, indicating promising biomarker properties in comparison to GAD65, a more established marker of beta cell injury. DCX potential was validated in vitro: chemically-induced necrosis of rat and human beta cells led to a discharge of intracellular doublecortin into the extracellular space, proportionate to the amount of injured cells, and similar to GAD65. In vivo, recombinant DCX showed favorable pharmacokinetic properties, with a half-life in plasma of around 3h. Combined, our findings provide first proof-of-principle for doublecortin as biomarker for beta cell injury in vitro, advocating its further validation as biomarker in vivo.
Collapse
|
16
|
Towards cytoprotection in the peritransplant period. Semin Immunol 2011; 23:209-13. [DOI: 10.1016/j.smim.2011.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 07/10/2011] [Indexed: 01/26/2023]
|
17
|
Tissue engineering on matrix: future of autologous tissue replacement. Semin Immunopathol 2011; 33:307-15. [PMID: 21279358 DOI: 10.1007/s00281-011-0258-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 01/19/2011] [Indexed: 01/03/2023]
Abstract
Tissue engineering aims at the creation of living neo-tissues identical or close to their native human counterparts. As basis of this approach, temporary biodegradable supporter matrices are fabricated in the shape of a desired construct, which promote tissue strength and provide functionality until sufficient neo-tissue is formed. Besides fully synthetic polymer-based scaffolds, decellularized biological tissue of xenogenic or homogenic origin can be used. In a second step, these scaffolds are seeded with autologous cells attaching to the scaffold microstructure. In order to promote neo-tissue formation and maturation, the seeded scaffolds are exposed to different forms of stimulation. In cardiovascular tissue engineering, this "conditioning" can be achieved via culture media and biomimetic in vitro exposure, e.g., using flow bioreactors. This aims at adequate cellular differentiation, proliferation, and extracellular matrix production to form a living tissue called the construct. These living autologous constructs, such as heart valves or vascular grafts, are created in vitro, comprising a viable interstitium with repair and remodeling capabilities already prior to implantation. In situ further in vivo remodeling is intended to recapitulate physiological vascular architecture and function. The remodeling mechanisms were shown to be dominated by monocytic infiltration and chemotactic host-cell attraction leading into a multifaceted inflammatory process and neo-tissue formation. Key molecules of these processes can be integrated into the scaffold matrix to direct cell and tissue fate in vivo.
Collapse
|