1
|
Asthana A, Chaimov D, Tamburrini R, Gazia C, Gallego A, Lozano T, Heo JH, Byers LN, Tomei A, Fraker CA, Walker SJ, Lee SJ, Opara EC, Orlando G. Decellularized human pancreatic extracellular matrix-based physiomimetic microenvironment for human islet culture. Acta Biomater 2023; 171:261-272. [PMID: 37742726 PMCID: PMC10615794 DOI: 10.1016/j.actbio.2023.09.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 09/26/2023]
Abstract
A strategy that seeks to combine the biophysical properties of inert encapsulation materials like alginate with the biochemical niche provided by pancreatic extracellular matrix (ECM)-derived biomaterials, could provide a physiomimetic pancreatic microenvironment for maintaining long-term islet viability and function in culture. Herein, we have demonstrated that incorporating human pancreatic decellularized ECM within alginate microcapsules results in a significant increase in Glucose Stimulation Index (GSI) and total insulin secreted by encapsulated human islets, compared to free islets and islets encapsulated in only alginate. ECM supplementation also resulted in long-term (58 days) maintenance of GSI levels, similar to that observed in free islets at the first time point (day 5). At early time points in culture, ECM promoted gene expression changes through ECM- and cell adhesion-mediated pathways, while it demonstrated a mitochondria-protective effect in the long-term. STATEMENT OF SIGNIFICANCE: The islet isolation process can damage the islet extracellular matrix, resulting in loss of viability and function. We have recently developed a detergent-free, DI-water based method for decellularization of human pancreas to produce a potent solubilized ECM. This ECM was added to alginate for microencapsulation of human islets, which resulted in significantly higher stimulation index and total insulin production, compared to only alginate capsules and free islets, over long-term culture. Using ECM to preserve islet health and function can improve transplantation outcomes, as well as provide novel materials and platforms for studying islet biology in microfluidic, organ-on-a-chip, bioreactor and 3D bioprinted systems.
Collapse
Affiliation(s)
- Amish Asthana
- Department of Surgery, Wake Forest Baptist Medical Center, Medical Center Boulevard, Winston Salem, USA; Wake Forest Institute for Regenerative Medicine, Winston Salem, USA; Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, USA.
| | - Deborah Chaimov
- Department of Surgery, Wake Forest Baptist Medical Center, Medical Center Boulevard, Winston Salem, USA; Wake Forest Institute for Regenerative Medicine, Winston Salem, USA; Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, USA
| | - Riccardo Tamburrini
- Department of Surgery, Wake Forest Baptist Medical Center, Medical Center Boulevard, Winston Salem, USA; Wake Forest Institute for Regenerative Medicine, Winston Salem, USA; Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, USA
| | - Carlo Gazia
- Department of Surgery, Wake Forest Baptist Medical Center, Medical Center Boulevard, Winston Salem, USA; Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, USA; Department of Surgery, Tor Vergata University of Rome, Italy
| | | | | | - Jun-Ho Heo
- Wake Forest Institute for Regenerative Medicine, Winston Salem, USA; Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Lori N Byers
- Department of Surgery, Wake Forest Baptist Medical Center, Medical Center Boulevard, Winston Salem, USA; Wake Forest Institute for Regenerative Medicine, Winston Salem, USA; Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, USA
| | - Alice Tomei
- Diabetes Research Institute, University of Miami, Miami, USA
| | | | - Stephen J Walker
- Wake Forest Institute for Regenerative Medicine, Winston Salem, USA; Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, USA
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Winston Salem, USA; Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, USA
| | - Emmanuel C Opara
- Wake Forest Institute for Regenerative Medicine, Winston Salem, USA; Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, USA
| | - Giuseppe Orlando
- Department of Surgery, Wake Forest Baptist Medical Center, Medical Center Boulevard, Winston Salem, USA; Wake Forest Institute for Regenerative Medicine, Winston Salem, USA; Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, USA.
| |
Collapse
|
2
|
Karacaer C, Demirci T, Cengiz H, Varim C, Tamer A. The effect of short-term intensive insulin therapy in newly-diagnosed Type-2 diabetic patients. Pak J Med Sci 2021; 37:1972-1978. [PMID: 34912428 PMCID: PMC8613043 DOI: 10.12669/pjms.37.7.4013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 06/02/2021] [Accepted: 06/12/2021] [Indexed: 11/16/2022] Open
Abstract
Objectives: We aimed to determine the effect of short-term intensive insulin therapy (SIIT) on long-term glycemic control in newly-diagnosed Type-2 diabetes mellitus (nT2DM) patients. Methods: In this retrospectively study conducted at Sakarya University Medical Faculty Training and Research Hospital Outpatient Clinic between 2016 and 2019, 65 nT2DM patients were enrolled soon after their SIIT was initiated and were followed for at least a year. Intensive insulin treatment was discontinued after three or 12 months in a total of 65 (23–73-year-old) patients who had been newly diagnosed with T2DM. Intensive insulin therapy was discontinued when glycemic control and the target Glycated Hemoglobin (HbA1c) level had been attained, after which oral anti-diabetic drug (OAD), long-term insulin, and diet therapies were pursued. Results: There was a significant decrease in mean HbA1c from 11.25±1.96% to 6.67±1.07%. Fasting plasma glucose (FPG) was found to be an independent predictor of whether intensive insulin therapy could be discontinued after three months in a model that included FPG, HbA1c, and body mass index measured at baseline. Patients with FPG>13.8 mmol/L were 7.6 times more likely to require intensive insulin therapy beyond three months. There were significant decreases in HbA1c and low-density lipoprotein-cholesterol concentration, but no change in C-peptide between baseline and 3 months of therapy. Conclusion: These results demonstrate that in nT2DM patients, intensive insulin therapy could be effective on long-term glycemic control and high FPG prior to three months of SIIT may predict poor long-term glycemic control.
Collapse
Affiliation(s)
- Cengiz Karacaer
- Dr. Cengiz Karacaer Department of Internal Medicine, Sakarya University Treatment and Research Hospital, Department of Internal Medicine, Sakarya, Turkey
| | - Taner Demirci
- Taner Demirci Assistant Professor, Department of Endocrinology and Metabolism, Sakarya University Treatment and Research Hospital, Department of Endocrinology and Metabolism, Sakarya, Turkey
| | - Hasret Cengiz
- Dr. Hasret Cengiz Department of Endocrinology and Metabolism, Sakarya University Treatment and Research Hospital, Department of Endocrinology and Metabolism, Sakarya, Turkey
| | - Ceyhun Varim
- Dr. Ceyhun Varim Department of Internal Medicine, Sakarya University Treatment and Research Hospital, Department of Internal Medicine, Sakarya, Turkey
| | - Ali Tamer
- Ali Tamer Professor, Department of Internal Medicine, Sakarya University Treatment and Research Hospital, Department of Internal Medicine, Sakarya, Turkey
| |
Collapse
|
3
|
Differential influence of tacrolimus and sirolimus on mitochondrial-dependent signaling for apoptosis in pancreatic cells. Mol Cell Biochem 2016; 418:91-102. [PMID: 27344165 DOI: 10.1007/s11010-016-2736-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/15/2016] [Indexed: 01/21/2023]
Abstract
To examine and compare the mitochondria-related cellular mechanisms by which tacrolimus (TAC) or sirolimus (SIR) immunosuppressive drugs alter the pancreatic exocrine and endocrine β-cell fate. Human exocrine PANC-1 and rat endocrine insulin-secreting RIN-m5F cells and isolated rat islets were submitted to 1-100 nM TAC or SIR. In cultures, insulin secretion was measured as endocrine cell function marker. Apoptosis was quantified by annexin 5 and propidium iodide staining. Cleaved caspase-3, Bax apoptosis indicators, and p53, p21 cell cycle regulators were detected by Western blot. Cell cycle and mitochondrial membrane potential (ΔΨm) were analyzed by flow cytometry and SA-beta-galactosidase (SA-β-gal) activity by fluorescence microscopy. Only TAC reduced insulin secretion by RIN-m5F after 24 h. TAC and SIR promoted moderate apoptosis in both PANC-1 and RIN-m5F after 24 h. Apoptosis was associated with up-regulated Bax (threefold) and cleaved caspase-3 (fivefold) but only in PANC-1, while p53 and p21 were up-regulated (twofold) in both cell lines. ΔΨm was impaired only in PANC-1 by TAC and SIR. Only SIR prompted cell cycle arrest in both cell lines. The induction of a premature senescence-like phenotype was confirmed in isolated islets by SA-β-gal activity. TAC and SIR are early inducers of pancreatic cell dysfunction and apoptosis but differentially alter endocrine and exocrine cells via mitochondrial-driven pathways. In rat islets, TAC and SIR prompt a senescence-like phenotype.
Collapse
|
4
|
Aly H, Rohatgi N, Marshall CA, Grossenheider TC, Miyoshi H, Stappenbeck TS, Matkovich SJ, McDaniel ML. A novel strategy to increase the proliferative potential of adult human β-cells while maintaining their differentiated phenotype. PLoS One 2013; 8:e66131. [PMID: 23776620 PMCID: PMC3680388 DOI: 10.1371/journal.pone.0066131] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 05/01/2013] [Indexed: 12/13/2022] Open
Abstract
Our previous studies demonstrated that Wnt/GSK-3/β-catenin and mTOR signaling are necessary to stimulate proliferative processes in adult human β-cells. Direct inhibition of GSK-3, that engages Wnt signaling downstream of the Wnt receptor, increases β-catenin nuclear translocation and β-cell proliferation but results in lower insulin content. Our current goal was to engage canonical and non-canonical Wnt signaling at the receptor level to significantly increase human β-cell proliferation while maintaining a β-cell phenotype in intact islets. We adopted a system that utilized conditioned medium from L cells that expressed Wnt3a, R-spondin-3 and Noggin (L-WRN conditioned medium). In addition we used a ROCK inhibitor (Y-27632) and SB-431542 (that results in RhoA inhibition) in these cultures. Treatment of intact human islets with L-WRN conditioned medium plus inhibitors significantly increased DNA synthesis ∼6 fold in a rapamycin-sensitive manner. Moreover, this treatment strikingly increased human β-cell proliferation ∼20 fold above glucose alone. Only the combination of L-WRN conditioned medium with RhoA/ROCK inhibitors resulted in substantial proliferation. Transcriptome-wide gene expression profiling demonstrated that L-WRN medium provoked robust changes in several signaling families, including enhanced β-catenin-mediated and β-cell-specific gene expression. This treatment also increased expression of Nr4a2 and Irs2 and resulted in phosphorylation of Akt. Importantly, glucose-stimulated insulin secretion and content were not downregulated by L-WRN medium treatment. Our data demonstrate that engaging Wnt signaling at the receptor level by this method leads to necessary crosstalk between multiple signaling pathways including activation of Akt, mTOR, Wnt/β-catenin, PKA/CREB, and inhibition of RhoA/ROCK that substantially increase human β-cell proliferation while maintaining the β-cell phenotype.
Collapse
Affiliation(s)
- Haytham Aly
- Department of Pathology and Immunology Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Nidhi Rohatgi
- Department of Pathology and Immunology Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Connie A. Marshall
- Department of Pathology and Immunology Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Tiffani C. Grossenheider
- Center for Pharmacogenomics, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Hiroyuki Miyoshi
- Department of Pathology and Immunology Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Thaddeus S. Stappenbeck
- Department of Pathology and Immunology Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Scot J. Matkovich
- Center for Pharmacogenomics, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Michael L. McDaniel
- Department of Pathology and Immunology Washington University in St. Louis, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|