1
|
Humardani FM, Endharti AT, Ningrum RA, Arsana Wiyasa IW, Mulyanata LT, Antonius Y, Jonathan J, Dwi Putra SE. Unique motif Sequences for early diagnosis of preeclampsia. Clin Chim Acta 2025; 574:120339. [PMID: 40348314 DOI: 10.1016/j.cca.2025.120339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025]
Abstract
Preeclampsia (PE) is a disease that significantly impacts both maternal and infant health with its prevalence varying across different ethnicities. Current diagnostic methods for PE typically identify the condition after 20 weeks of gestation, often when the disease has already manifested and reached an advanced stage. The situation underscores the urgent need for early biomarkers capable of effective screening and diagnosis. Our review addresses this challenge by utilizing bioinformatics approaches as an alternative method prior to preclinical and clinical studies. Specifically, we focus on FRAGmentomics-based Methylation Analysis (FRAGMA), targeting the CGCGCGG sequence motif for methylation studies in cell-free DNA (cfDNA). Since cfDNA is largely derived from the placenta, the FRAGMA approach is particularly promising, given that the primary pathophysiology of PE originates in the placenta, and methylation patterns are unique to specific tissues. In the previous research, we identified 66 genes containing this sequence motif that are implicated in the pathophysiology of PE, and only six genes - FN1, ITGA2, ITGA5, ITGB1, ITGB3, and VWF - show potential as early detection biomarkers for PE. These genes still require further investigation to confirm their utility as biomarkers for PE in the future studies.
Collapse
Affiliation(s)
| | - Agustina Tri Endharti
- Doctoral Program in Medical Science, Faculty of Medicine Universitas Brawijaya, Malang, Indonesia.
| | - Ratih Asmana Ningrum
- Research Center for Genetic Engineering, National Research and Innovation Agency, Cibinong, Bogor, Indonesia.
| | - I Wayan Arsana Wiyasa
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia.
| | | | - Yulanda Antonius
- Faculty of Biotechnology, University of Surabaya, Surabaya, Indonesia.
| | - Jonathan Jonathan
- Faculty of Biotechnology, University of Surabaya, Surabaya, Indonesia.
| | | |
Collapse
|
2
|
Olaniru OE, Toczyska K, Guccio N, Giera S, Piao X, King AJF, Jones PM, Persaud SJ. Spatiotemporal profiling of adhesion G protein-coupled receptors in developing mouse and human pancreas reveals a role for GPR56 in islet development. Cell Mol Life Sci 2025; 82:129. [PMID: 40137991 PMCID: PMC11947406 DOI: 10.1007/s00018-025-05659-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025]
Abstract
INTRODUCTION G protein-coupled receptors (GPCRs) are cell-surface proteins that are targeted therapeutically for a range of disorders, including diabetes. Adhesion GPCRs (aGPCRs) are the second largest class of the GPCR superfamily and some members of this family have been implicated in appropriate organ development. However, the role of aGPCRs in endocrine pancreas specification is not yet known. METHODS Here, we systematically characterised expression of mRNAs encoding aGPCRs and their ligands in developing mouse and human pancreas using our own and publicly available single-cell RNA sequencing and spatial transcriptomics data, and we conducted qPCR analysis of aGPCR expression in human pancreas at different gestational stages. We then investigated the role of GPR56 (ADGRG1), the most abundant aGPCR in pancreatic endocrine progenitors, in islet development using Gpr56 null mice and their wildtype littermates. RESULTS We demonstrated that aGPCRs are dynamically expressed during mouse and human pancreas development, with specific aGPCR mRNAs expressed in distinct endocrine, endothelial, mesenchymal, acinar, ductal, and immune cell clusters. aGPCR ligand mRNAs were mostly expressed by non-endocrine cells, and the most highly expressed receptor-ligand interacting mRNA pairs were those encoding GPR56 and COL3A1. Deletion of Gpr56 in neonatal mice was associated with an altered α-/β-/δ-cell ratio and reduced β-cell proliferation. CONCLUSION Our data show that aGPCRs are expressed at key stages of human and mouse pancreas endocrine lineage decisions, and analysis of pancreases from Gpr56 knockout mice implicate this aGPCR in the development of a full complement of β-cells.
Collapse
Affiliation(s)
- Oladapo E Olaniru
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Klaudia Toczyska
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Nunzio Guccio
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Stefanie Giera
- Department of Medicine, Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Xianhua Piao
- Department of Medicine, Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Pediatrics, University of California at San Francisco, San Francisco, CA, USA
| | - Aileen J F King
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Peter M Jones
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Shanta J Persaud
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Guy's Campus, London, SE1 1UL, UK.
| |
Collapse
|
3
|
Endo Kumata Y, Inagaki A, Nakamura Y, Imura T, Saito R, Katano T, Suzuki S, Tokodai K, Kamei T, Unno M, Watanabe K, Tabata Y, Goto M. A novel method of pancreatic islet transplantation at the liver surface using a gelatin hydrogel nonwoven fabric. Cell Transplant 2025; 34:9636897251328419. [PMID: 40264358 PMCID: PMC12035123 DOI: 10.1177/09636897251328419] [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: 11/10/2024] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 04/24/2025] Open
Abstract
Considering the limitations of intraportal transplantation (Tx), we sought to establish an alternative approach for it-transplanting islets onto the liver surface (LS) by optimizing adipose tissue-derived stem cell (ADSC) co-Tx procedures with a gelatin hydrogel nonwoven fabric (GHNF). In the in vivo study, we examined the use of the GHNF, the effectiveness of islet covering materials, and preferred procedures for ADSC co-Tx using a syngeneic rat model. Immunohistochemical staining was performed to evaluate the extracellular matrix (ECM) expression and angiogenesis. In the in vitro study, we analyzed the culture supernatants to identify crucial factors secreted from ADSCs in different ADSC co-Tx procedures. It was shown that the GHNF should be used to cover the islets but not to embed internally (encapsulate) them. Utilization of the GHNF in LS Tx resulted in significantly better glucose changes (P = 0.0002) and cure rate of diabetic recipients (P = 0.0003) than the use of a common adhesion barrier. Although neovascularization was comparable among groups, ECM reconstitution tended to be higher when the GHNF was used. ADSC co-Tx further enhanced ECM reconstitution only when ADSCs were cultured in the GHNF before islet Tx. Leptin, vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), and several chemokines were identified as candidate factors for enhancing ECM reconstitution (P < 0.001). The inhibition assay using antagonist suggested that leptin might be at least in part responsible for the difference in transplant efficiency in distinct ADSC co-Tx methods. This study showed that the GHNF effectively improved the outcomes of LS islet Tx, mainly due to ECM reconstitution around the islets. Furthermore, we established a novel method of LS islet Tx by combining a GHNF with ADSCs, which is equally effective as intraportal Tx.
Collapse
Affiliation(s)
- Yukiko Endo Kumata
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akiko Inagaki
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiro Nakamura
- Division of Pathology, Graduate School of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Takehiro Imura
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryusuke Saito
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takumi Katano
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shoki Suzuki
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuaki Tokodai
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Kamei
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kimiko Watanabe
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiko Tabata
- Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masafumi Goto
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
4
|
Ballesteros-Pla C, Sevillano J, Sánchez-Alonso MG, Limones M, Pita J, Zapatería B, Sanz-Cuadrado MI, Pizarro-Delgado J, Izquierdo-Lahuerta A, Medina-Gómez G, Herradón G, Ramos-Álvarez MDP. Constitutive Pleiotrophin Deletion Results in a Phenotype with an Altered Pancreatic Morphology and Function in Old Mice. Int J Mol Sci 2024; 25:10960. [PMID: 39456743 PMCID: PMC11507919 DOI: 10.3390/ijms252010960] [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: 09/16/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Pleiotrophin (PTN) is crucial for embryonic development and pancreas organogenesis as it regulates metainflammation, metabolic homeostasis, thermogenesis, and glucose tolerance. Pleiotrophin deletion is associated with a lipodystrophic phenotype in which adipose tissue plasticity is altered in late life. This study explored the impact of pleiotrophin deletion on pancreatic morphology and function in later life. We analyzed glucose tolerance and circulating parameters on female wild-type (Ptn+/+) and knock-out (Ptn-/-) mice. At 9 and 15 months, we conducted morphometric analyses of pancreatic islets and evaluated the levels of insulin, glucagon, somatostatin, glucose transporter 2 (GLUT2), vesicle-associated membrane protein 2 (VAMP2), and synaptosome-associated protein 25 (SNAP25) via immunofluorescence. The effect of PTN on glucose-stimulated insulin secretion (GSIS) was evaluated in INS1E cells and isolated islets. Ptn-/- mice showed hyperinsulinemia, impaired glucose tolerance, and increased homeostatic model assessment for insulin resistance (HOMA-IR) with age. While Ptn+/+ islets enlarge with age, in Ptn-/- mice, the median size decreased, and insulin content increased. Vesicle transport and exocytosis proteins were significantly increased in 9-month-old Ptn-/- islets. Islets from Ptn-/- mice showed impaired GSIS and decreased cell membrane localization of GLUT2 whereas, PTN increased GSIS in INS1E cells. Ptn deletion accelerated age-related changes in the endocrine pancreas, affecting islet number and size, and altering VAMP2 and SNAP25 levels and GLUT2 localization leading to impaired GSIS and insulin accumulation in islets.
Collapse
Affiliation(s)
- Cristina Ballesteros-Pla
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660 Madrid, Spain; (C.B.-P.); (J.S.); (M.G.S.-A.); (M.L.); (J.P.); (B.Z.); (M.I.S.-C.); (J.P.-D.)
| | - Julio Sevillano
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660 Madrid, Spain; (C.B.-P.); (J.S.); (M.G.S.-A.); (M.L.); (J.P.); (B.Z.); (M.I.S.-C.); (J.P.-D.)
| | - María Gracia Sánchez-Alonso
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660 Madrid, Spain; (C.B.-P.); (J.S.); (M.G.S.-A.); (M.L.); (J.P.); (B.Z.); (M.I.S.-C.); (J.P.-D.)
| | - María Limones
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660 Madrid, Spain; (C.B.-P.); (J.S.); (M.G.S.-A.); (M.L.); (J.P.); (B.Z.); (M.I.S.-C.); (J.P.-D.)
| | - Jimena Pita
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660 Madrid, Spain; (C.B.-P.); (J.S.); (M.G.S.-A.); (M.L.); (J.P.); (B.Z.); (M.I.S.-C.); (J.P.-D.)
| | - Begoña Zapatería
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660 Madrid, Spain; (C.B.-P.); (J.S.); (M.G.S.-A.); (M.L.); (J.P.); (B.Z.); (M.I.S.-C.); (J.P.-D.)
- Department of Medicine, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Marta Inmaculada Sanz-Cuadrado
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660 Madrid, Spain; (C.B.-P.); (J.S.); (M.G.S.-A.); (M.L.); (J.P.); (B.Z.); (M.I.S.-C.); (J.P.-D.)
| | - Javier Pizarro-Delgado
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660 Madrid, Spain; (C.B.-P.); (J.S.); (M.G.S.-A.); (M.L.); (J.P.); (B.Z.); (M.I.S.-C.); (J.P.-D.)
| | - Adriana Izquierdo-Lahuerta
- Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, 28922 Madrid, Spain; (A.I.-L.); (G.M.-G.)
| | - Gema Medina-Gómez
- Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, 28922 Madrid, Spain; (A.I.-L.); (G.M.-G.)
| | - Gonzalo Herradón
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte Urbanización Montepríncipe, 28660 Madrid, Spain;
| | - María del Pilar Ramos-Álvarez
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660 Madrid, Spain; (C.B.-P.); (J.S.); (M.G.S.-A.); (M.L.); (J.P.); (B.Z.); (M.I.S.-C.); (J.P.-D.)
| |
Collapse
|
5
|
Poon F, Sambathkumar R, Korytnikov R, Aghazadeh Y, Oakie A, Misra PS, Sarangi F, Nostro MC. Tankyrase inhibition promotes endocrine commitment of hPSC-derived pancreatic progenitors. Nat Commun 2024; 15:8754. [PMID: 39384787 PMCID: PMC11464881 DOI: 10.1038/s41467-024-53068-w] [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: 06/27/2022] [Accepted: 09/27/2024] [Indexed: 10/11/2024] Open
Abstract
Human pluripotent stem cells (hPSCs) have the potential to differentiate into various cell types, including pancreatic insulin-producing β cells, which are crucial for developing therapies for diabetes. However, current methods for directing hPSC differentiation towards pancreatic β-like cells are often inefficient and produce cells that do not fully resemble the native counterparts. Here, we report that highly selective tankyrase inhibitors, such as WIKI4, significantly enhances pancreatic differentiation from hPSCs. Our results show that WIKI4 promotes the formation of pancreatic progenitors that give rise to islet-like cells with improved β-like cell frequencies and glucose responsiveness compared to our standard cultures. These findings not only advance our understanding of pancreatic development, but also provide a promising new tool for generating pancreatic cells for research and potential therapeutic applications.
Collapse
Affiliation(s)
- Frankie Poon
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Sana Biotechnology, 300 Technology Square, Cambridge, MA, 02139, USA
| | - Rangarajan Sambathkumar
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
- Allarta Life Science Inc., 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - Roman Korytnikov
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Yasaman Aghazadeh
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
- Montreal Clinical Research Institute (IRCM), University of Montreal, Department of Medicine, Montreal, H2W 1R7, QC, Canada
| | - Amanda Oakie
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Paraish S Misra
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Department of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Farida Sarangi
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - M Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
6
|
Santos da Silva T, da Silva-Júnior LN, Horvath-Pereira BDO, Valbão MCM, Garcia MHH, Lopes JB, Reis CHB, Barreto RDSN, Buchaim DV, Buchaim RL, Miglino MA. The Role of the Pancreatic Extracellular Matrix as a Tissue Engineering Support for the Bioartificial Pancreas. Biomimetics (Basel) 2024; 9:598. [PMID: 39451804 PMCID: PMC11505355 DOI: 10.3390/biomimetics9100598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/22/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic condition primarily managed with insulin replacement, leading to significant treatment costs. Complications include vasculopathy, cardiovascular diseases, nephropathy, neuropathy, and reticulopathy. Pancreatic islet transplantation is an option but its success does not depend solely on adequate vascularization. The main limitations to clinical islet transplantation are the scarcity of human pancreas, the need for immunosuppression, and the inadequacy of the islet isolation process. Despite extensive research, T1DM remains a major global health issue. In 2015, diabetes affected approximately 415 million people, with projected expenditures of USD 1.7 trillion by 2030. Pancreas transplantation faces challenges due to limited organ availability and complex vascularization. T1DM is caused by the autoimmune destruction of insulin-producing pancreatic cells. Advances in biomaterials, particularly the extracellular matrix (ECM), show promise in tissue reconstruction and transplantation, offering structural and regulatory functions critical for cell migration, differentiation, and adhesion. Tissue engineering aims to create bioartificial pancreases integrating insulin-producing cells and suitable frameworks. This involves decellularization and recellularization techniques to develop biological scaffolds. The challenges include replicating the pancreas's intricate architecture and maintaining cell viability and functionality. Emerging technologies, such as 3D printing and advanced biomaterials, have shown potential in constructing bioartificial organs. ECM components, including collagens and glycoproteins, play essential roles in cell adhesion, migration, and differentiation. Clinical applications focus on developing functional scaffolds for transplantation, with ongoing research addressing immunological responses and long-term efficacy. Pancreatic bioengineering represents a promising avenue for T1DM treatment, requiring further research to ensure successful implementation.
Collapse
Affiliation(s)
- Thamires Santos da Silva
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil; (T.S.d.S.); (L.N.d.S.-J.); (B.d.O.H.-P.); (R.d.S.N.B.); (D.V.B.); (R.L.B.)
| | - Leandro Norberto da Silva-Júnior
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil; (T.S.d.S.); (L.N.d.S.-J.); (B.d.O.H.-P.); (R.d.S.N.B.); (D.V.B.); (R.L.B.)
- Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil; (M.C.M.V.); (M.H.H.G.); (J.B.L.)
| | - Bianca de Oliveira Horvath-Pereira
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil; (T.S.d.S.); (L.N.d.S.-J.); (B.d.O.H.-P.); (R.d.S.N.B.); (D.V.B.); (R.L.B.)
| | - Maria Carolina Miglino Valbão
- Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil; (M.C.M.V.); (M.H.H.G.); (J.B.L.)
| | | | - Juliana Barbosa Lopes
- Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil; (M.C.M.V.); (M.H.H.G.); (J.B.L.)
| | - Carlos Henrique Bertoni Reis
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil;
- UNIMAR Beneficent Hospital (HBU), Medical School, University of Marilia (UNIMAR), Marilia 17525-160, Brazil
| | - Rodrigo da Silva Nunes Barreto
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil; (T.S.d.S.); (L.N.d.S.-J.); (B.d.O.H.-P.); (R.d.S.N.B.); (D.V.B.); (R.L.B.)
- Department of Animal Morphology and Physiology, Faculty of Agricultural and Veterinary Sciences, São Paulo State University, Jaboticabal 14884-900, Brazil
| | - Daniela Vieira Buchaim
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil; (T.S.d.S.); (L.N.d.S.-J.); (B.d.O.H.-P.); (R.d.S.N.B.); (D.V.B.); (R.L.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil;
- Medical School, University Center of Adamantina (UNIFAI), Adamantina 17800-000, Brazil
| | - Rogerio Leone Buchaim
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil; (T.S.d.S.); (L.N.d.S.-J.); (B.d.O.H.-P.); (R.d.S.N.B.); (D.V.B.); (R.L.B.)
- Department of Biological Sciences, Bauru School of Dentistry (FOB/USP), University of Sao Paulo, Bauru 17012-901, Brazil
| | - Maria Angelica Miglino
- Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil; (M.C.M.V.); (M.H.H.G.); (J.B.L.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil;
- Postgraduate Program in Animal Health, Production and Environment, University of Marilia (UNIMAR), Marilia 17525-902, Brazil
| |
Collapse
|
7
|
Ajoolabady A, Pratico D, Ren J. Endothelial dysfunction: mechanisms and contribution to diseases. Acta Pharmacol Sin 2024; 45:2023-2031. [PMID: 38773228 PMCID: PMC11420364 DOI: 10.1038/s41401-024-01295-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 04/16/2024] [Indexed: 05/23/2024]
Abstract
The endothelium, lining the inner surface of blood vessels and spanning approximately 3 m2, serves as the largest organ in the body. Comprised of endothelial cells, the endothelium interacts with other bodily components including the bloodstream, circulating cells, and the lymphatic system. Functionally, the endothelium primarily synchronizes vascular tone (by balancing vasodilation and vasoconstriction) and prevents vascular inflammation and pathologies. Consequently, endothelial dysfunction disrupts vascular homeostasis, leading to vascular injuries and diseases such as cardiovascular, cerebral, and metabolic diseases. In this opinion/perspective piece, we explore the recently identified mechanisms of endothelial dysfunction across various disease subsets and critically evaluate the strengths and limitations of current therapeutic interventions at the pre-clinical level.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| |
Collapse
|
8
|
Doherty DT, Khambalia HA, van Dellen D, Jennings RE, Piper Hanley K. Unlocking the post-transplant microenvironment for successful islet function and survival. Front Endocrinol (Lausanne) 2023; 14:1250126. [PMID: 37711891 PMCID: PMC10497759 DOI: 10.3389/fendo.2023.1250126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/07/2023] [Indexed: 09/16/2023] Open
Abstract
Islet transplantation (IT) offers the potential to restore euglycemia for patients with type 1 diabetes mellitus (T1DM). Despite improvements in islet isolation techniques and immunosuppressive regimes, outcomes remain suboptimal with UK five-year graft survivals (5YGS) of 55% and most patients still requiring exogenous insulin after multiple islet infusions. Native islets have a significant non-endocrine component with dense extra-cellular matrix (ECM), important for islet development, cell survival and function. Collagenase isolation necessarily disrupts this complex islet microenvironment, leaving islets devoid of a supporting framework and increasing vulnerability of transplanted islets. Following portal venous transplantation, a liver injury response is potentially induced, which typically results in inflammation and ECM deposition from liver specific myofibroblasts. The impact of this response may have important impact on islet survival and function. A fibroblast response and ECM deposition at the kidney capsule and eye chamber alongside other implantation sites have been shown to be beneficial for survival and function. Investigating the implantation site microenvironment and the interactions of transplanted islets with ECM proteins may reveal therapeutic interventions to improve IT and stem-cell derived beta-cell therapy.
Collapse
Affiliation(s)
- Daniel T. Doherty
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom
- Department of Renal & Pancreatic Transplantation, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Hussein A. Khambalia
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom
- Department of Renal & Pancreatic Transplantation, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - David van Dellen
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom
- Department of Renal & Pancreatic Transplantation, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Rachel E. Jennings
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom
- Department of Endocrinology, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Karen Piper Hanley
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
9
|
Tixi W, Maldonado M, Chang YT, Chiu A, Yeung W, Parveen N, Nelson MS, Hart R, Wang S, Hsu WJ, Fueger P, Kopp JL, Huising MO, Dhawan S, Shih HP. Coordination between ECM and cell-cell adhesion regulates the development of islet aggregation, architecture, and functional maturation. eLife 2023; 12:e90006. [PMID: 37610090 PMCID: PMC10482429 DOI: 10.7554/elife.90006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/12/2023] [Indexed: 08/24/2023] Open
Abstract
Pancreatic islets are three-dimensional cell aggregates consisting of unique cellular composition, cell-to-cell contacts, and interactions with blood vessels. Cell aggregation is essential for islet endocrine function; however, it remains unclear how developing islets establish aggregation. By combining genetic animal models, imaging tools, and gene expression profiling, we demonstrate that islet aggregation is regulated by extracellular matrix signaling and cell-cell adhesion. Islet endocrine cell-specific inactivation of extracellular matrix receptor integrin β1 disrupted blood vessel interactions but promoted cell-cell adhesion and the formation of larger islets. In contrast, ablation of cell-cell adhesion molecule α-catenin promoted blood vessel interactions yet compromised islet clustering. Simultaneous removal of integrin β1 and α-catenin disrupts islet aggregation and the endocrine cell maturation process, demonstrating that establishment of islet aggregates is essential for functional maturation. Our study provides new insights into understanding the fundamental self-organizing mechanism for islet aggregation, architecture, and functional maturation.
Collapse
Affiliation(s)
- Wilma Tixi
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Maricela Maldonado
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
- Department of Biomedical Engineering, College of Engineering, California State University, Long BeachLong BeachUnited States
| | - Ya-Ting Chang
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Amy Chiu
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Wilson Yeung
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Nazia Parveen
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Michael S Nelson
- Light Microscopy Core, Beckman Research Institute, City of HopeDuarteUnited States
| | - Ryan Hart
- Department of Neurobiology, Physiology and Behavior, University of California, DavisDavisUnited States
| | - Shihao Wang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British ColumbiaVancouverCanada
| | - Wu Jih Hsu
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British ColumbiaVancouverCanada
| | - Patrick Fueger
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Janel L Kopp
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British ColumbiaVancouverCanada
| | - Mark O Huising
- Department of Neurobiology, Physiology and Behavior, University of California, DavisDavisUnited States
- Department of Physiology and Membrane Biology, School of Medicine, University of California, DavisDavisUnited States
| | - Sangeeta Dhawan
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Hung Ping Shih
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| |
Collapse
|
10
|
Ballesteros-Pla C, Sánchez-Alonso MG, Pizarro-Delgado J, Zuccaro A, Sevillano J, Ramos-Álvarez MP. Pleiotrophin and metabolic disorders: insights into its role in metabolism. Front Endocrinol (Lausanne) 2023; 14:1225150. [PMID: 37484951 PMCID: PMC10360176 DOI: 10.3389/fendo.2023.1225150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/26/2023] [Indexed: 07/25/2023] Open
Abstract
Pleiotrophin (PTN) is a cytokine which has been for long studied at the level of the central nervous system, however few studies focus on its role in the peripheral organs. The main aim of this review is to summarize the state of the art of what is known up to date about pleiotrophin and its implications in the main metabolic organs. In summary, pleiotrophin promotes the proliferation of preadipocytes, pancreatic β cells, as well as cells during the mammary gland development. Moreover, this cytokine is important for the structural integrity of the liver and the neuromuscular junction in the skeletal muscle. From a metabolic point of view, pleiotrophin plays a key role in the maintenance of glucose and lipid as well as whole-body insulin homeostasis and favors oxidative metabolism in the skeletal muscle. All in all, this review proposes pleiotrophin as a druggable target to prevent from the development of insulin-resistance-related pathologies.
Collapse
|
11
|
Abstract
The islets of Langerhans are highly organized structures that have species-specific, three-dimensional tissue architecture. Islet architecture is critical for proper hormone secretion in response to nutritional stimuli. Islet architecture is disrupted in all types of diabetes mellitus and in cadaveric islets for transplantation during isolation, culture, and perfusion, limiting patient outcomes. Moreover, recapitulating native islet architecture remains a key challenge for in vitro generation of islets from stem cells. In this review, we discuss work that has led to the current understanding of determinants of pancreatic islet architecture, and how this architecture is maintained or disrupted during tissue remodeling in response to normal and pathological metabolic changes. We further discuss both empirical and modeling data that highlight the importance of islet architecture for islet function.
Collapse
Affiliation(s)
- Melissa T. Adams
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Barak Blum
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
- CONTACT Barak Blum Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI53705, USA
| |
Collapse
|
12
|
Sevillano J, Liang A, Strutt B, Hill TG, Szlapinski S, Ramos-Álvarez MP, Hill DJ. Pleiotrophin Expression and Actions in Pancreatic β-Cells. Front Endocrinol (Lausanne) 2022; 13:777868. [PMID: 35250852 PMCID: PMC8894601 DOI: 10.3389/fendo.2022.777868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/06/2022] [Indexed: 11/13/2022] Open
Abstract
Pleiotrophin (PTN) is a heparin-binding cytokine that is widely expressed during early development and increases in maternal circulation during pregnancy.Aged PTN-deficient mice exhibit insulin resistance, suggesting a role in metabolic control. The objectives of this study were to determine if PTN is expressed in mouse pancreatic β-cells in young vs. adult animals, and its effects on DNA synthesis, β-cell gene expression and glucose-stimulated insulin secretion (GSIS). The Ptn gene was expressed in isolated fractions of young mouse β-cells, especially within immature β-cells with low glucose transporter 2 expression. Expression was retained in the adult pancreas but did not significantly change during pregnancy. PTN and its receptor, phosphotyrosine phosphatase-β/ζ, were also expressed in the proliferative INS1E β-cell line. Fluorescence immunohistochemistry showed that PTN peptide was present in islets of Langerhans in adult mice, associated predominantly with β-cells. The percentage of β-cells staining for PTN did not alter during mouse pregnancy, but intense staining was seen during β-cell regeneration in young mice following depletion of β-cells with streptozotocin. Incubation of INS1E cells with PTN resulted in an increased DNA synthesis as measured by Ki67 localization and increased expression of Pdx1 and insulin. However, both DNA synthesis and GSIS were not altered by PTN in isolated adult mouse islets. The findings show that Ptn is expressed in mouse β-cells in young and adult life and could potentially contribute to adaptive increases in β-cell mass during early life or pregnancy.
Collapse
Affiliation(s)
- Julio Sevillano
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad CEU San Pablo, CEU Universities, Madrid, Spain
- Lawson Health Research Institute, St Joseph Health Care, London, ON, Canada
| | - Aileen Liang
- Lawson Health Research Institute, St Joseph Health Care, London, ON, Canada
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Brenda Strutt
- Lawson Health Research Institute, St Joseph Health Care, London, ON, Canada
| | - Thomas G. Hill
- Lawson Health Research Institute, St Joseph Health Care, London, ON, Canada
| | - Sandra Szlapinski
- Lawson Health Research Institute, St Joseph Health Care, London, ON, Canada
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Maria Pilar Ramos-Álvarez
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad CEU San Pablo, CEU Universities, Madrid, Spain
| | - David J. Hill
- Lawson Health Research Institute, St Joseph Health Care, London, ON, Canada
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Department of Medicine, Western University, London, ON, Canada
- *Correspondence: David J. Hill,
| |
Collapse
|
13
|
Shojaie L, Rahimi Y, Zolbin MM, Daghigh F, Kajbafzadeh AM. Characterization Methods of Acellularized Tissue and Organs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1345:1-6. [PMID: 34582009 DOI: 10.1007/978-3-030-82735-9_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
The extracellular matrix (ECM) of mammalian organs and tissues has been applied as a substitute scaffold to simplify the restoration and reconstruction of several tissues. Such scaffolds are prepared in various arrangements including sheets, powders, and hydrogels. One of the more applicable processes is using natural scaffolds, for this purpose discarded tissues or organs are naturally derived by processes that comprised decellularization of following tissues or organs. Protection of the complex structure and 3D (three dimensional) ultrastructure of the ECM is extremely necessary but it is predictable that all protocols of decellularization end in disruption of the architecture and potential loss of surface organization and configuration. Tissue decellularization with conservation of ECM bioactivity and integrity can be improved by providing well-designed protocols regarding the agents and decellularization techniques operated during processing. An overview of the characterization of decellularized scaffolds and the role of reagnets can validate the applied methods' efficacy.
Collapse
Affiliation(s)
- Layla Shojaie
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran.,Department of Medicine Division of GI/Liver Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Yekta Rahimi
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Majidi Zolbin
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Faezeh Daghigh
- Department of Physiology, Tabriz Branch, Islamic Azad University, Tabriz, Iran.,Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abdol-Mohammad Kajbafzadeh
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran. .,, No. 62, Dr. Gharib's Street, Keshavarz Boulevard, (PANNEK, #6), 1419433151, Tehran, Iran.
| |
Collapse
|
14
|
Wiedenmann S, Breunig M, Merkle J, von Toerne C, Georgiev T, Moussus M, Schulte L, Seufferlein T, Sterr M, Lickert H, Weissinger SE, Möller P, Hauck SM, Hohwieler M, Kleger A, Meier M. Single-cell-resolved differentiation of human induced pluripotent stem cells into pancreatic duct-like organoids on a microwell chip. Nat Biomed Eng 2021; 5:897-913. [PMID: 34239116 PMCID: PMC7611572 DOI: 10.1038/s41551-021-00757-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 06/01/2021] [Indexed: 02/06/2023]
Abstract
Creating in vitro models of diseases of the pancreatic ductal compartment requires a comprehensive understanding of the developmental trajectories of pancreas-specific cell types. Here we report the single-cell characterization of the differentiation of pancreatic duct-like organoids (PDLOs) from human induced pluripotent stem cells (hiPSCs) on a microwell chip that facilitates the uniform aggregation and chemical induction of hiPSC-derived pancreatic progenitors. Using time-resolved single-cell transcriptional profiling and immunofluorescence imaging of the forming PDLOs, we identified differentiation routes from pancreatic progenitors through ductal intermediates to two types of mature duct-like cells and a few non-ductal cell types. PDLO subpopulations expressed either mucins or the cystic fibrosis transmembrane conductance regulator, and resembled human adult duct cells. We also used the chip to uncover ductal markers relevant to pancreatic carcinogenesis, and to establish PDLO co-cultures with stellate cells, which allowed for the study of epithelial-mesenchymal signalling. The PDLO microsystem could be used to establish patient-specific pancreatic duct models.
Collapse
Affiliation(s)
- Sandra Wiedenmann
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Ingolstaedter Landstraße 1, 85764 Neuherberg, Germany
| | - Markus Breunig
- Department of Internal Medicine I, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Jessica Merkle
- Department of Internal Medicine I, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Christine von Toerne
- Research Unit Protein Science, Helmholtz Zentrum München, Heidemannstraße 1, 80939 Müunich, Germany
| | - Tihomir Georgiev
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Ingolstaedter Landstraße 1, 85764 Neuherberg, Germany
| | - Michel Moussus
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Ingolstaedter Landstraße 1, 85764 Neuherberg, Germany
| | - Lucas Schulte
- Department of Internal Medicine I, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Thomas Seufferlein
- Department of Internal Medicine I, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Michael Sterr
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Ingolstaedter Landstraße 1, 85764 Neuherberg, Germany,German Center for Diabetes Research (DZD), Ingolstaedter Landstraße 1, 85764 Neuherberg, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Ingolstaedter Landstraße 1, 85764 Neuherberg, Germany,German Center for Diabetes Research (DZD), Ingolstaedter Landstraße 1, 85764 Neuherberg, Germany,Institute of Stem Cell Research, Helmholtz Zentrum München, Ingolstaedter Landstraße 1, 85764 Neuherberg, Germany,Technical University of Munich, School of Medicine, Ismaninger Straße 22, 81675 Munich, Germany
| | | | - Peter Möller
- Institute for Pathology, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Stefanie M. Hauck
- Research Unit Protein Science, Helmholtz Zentrum München, Heidemannstraße 1, 80939 Müunich, Germany
| | - Meike Hohwieler
- Department of Internal Medicine I, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany,Corresponding authors: ; ;
| | - Alexander Kleger
- Department of Internal Medicine I, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany,Corresponding authors: ; ;
| | - Matthias Meier
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Ingolstaedter Landstraße 1, 85764 Neuherberg, Germany,Technical University of Munich, School of Medicine, Ismaninger Straße 22, 81675 Munich, Germany,Corresponding authors: ; ;
| |
Collapse
|
15
|
Murray HE, Zafar A, Qureshi KM, Paget MB, Bailey CJ, Downing R. The potential role of multifunctional human amniotic epithelial cells in pancreatic islet transplantation. J Tissue Eng Regen Med 2021; 15:599-611. [PMID: 34216434 DOI: 10.1002/term.3214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/23/2021] [Indexed: 11/08/2022]
Abstract
Pancreatic islet cell transplantation has proven efficacy as a treatment for type 1 diabetes mellitus, chiefly in individuals who are refractory to conventional insulin replacement therapy. At present its clinical use is restricted, firstly by the limited access to suitable donor organs but also due to factors associated with the current clinical transplant procedure which inadvertently impair the long-term functionality of the islet graft. Of note, the physical, biochemical, inflammatory, and immunological stresses to which islets are subjected, either during pretransplant processing or following implantation are detrimental to their sustained viability, necessitating repeated islet infusions to attain adequate glucose control. Progressive decline in functional beta (β)-cell mass leads to graft failure and the eventual re-instatement of exogenous insulin treatment. Strategies which protect and/or preserve optimal islet function in the peri-transplant period would improve clinical outcomes. Human amniotic epithelial cells (HAEC) exhibit both pluripotency and immune-privilege and are ideally suited for use in replacement and regenerative therapies. The HAEC secretome exhibits trophic, anti-inflammatory, and immunomodulatory properties of relevance to islet graft survival. Facilitated by β-cell supportive 3D cell culture systems, HAEC may be integrated with islets bringing them into close spatial arrangement where they may exert paracrine influences that support β-cell function, reduce hypoxia-induced islet injury, and alter islet alloreactivity. The present review details the potential of multifunctional HAEC in the context of islet transplantation, with a focus on the innate capabilities that may counter adverse events associated with the current clinical transplant protocol to achieve long-term islet graft function.
Collapse
Affiliation(s)
- Hilary E Murray
- The Islet Research Laboratory, Worcester Clinical Research Unit, Worcestershire Acute Hospitals NHS Trust, Worcester, UK
| | - Ali Zafar
- The Islet Research Laboratory, Worcester Clinical Research Unit, Worcestershire Acute Hospitals NHS Trust, Worcester, UK.,Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Khalid M Qureshi
- The Islet Research Laboratory, Worcester Clinical Research Unit, Worcestershire Acute Hospitals NHS Trust, Worcester, UK.,Bradford Royal Infirmary, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Michelle B Paget
- The Islet Research Laboratory, Worcester Clinical Research Unit, Worcestershire Acute Hospitals NHS Trust, Worcester, UK
| | - Clifford J Bailey
- Diabetes Research, School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Richard Downing
- The Islet Research Laboratory, Worcester Clinical Research Unit, Worcestershire Acute Hospitals NHS Trust, Worcester, UK
| |
Collapse
|
16
|
Schiesser JV, Loudovaris T, Thomas HE, Elefanty AG, Stanley EG. Integrin αvβ5 heterodimer is a specific marker of human pancreatic beta cells. Sci Rep 2021; 11:8315. [PMID: 33859325 PMCID: PMC8050092 DOI: 10.1038/s41598-021-87805-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/26/2021] [Indexed: 11/09/2022] Open
Abstract
The identification of cell surface markers specific to pancreatic beta cells is important for both the study of islet biology and for investigating the pathophysiology of diseases in which this cell type is lost or damaged. Following analysis of publicly available RNAseq data, we identified specific integrin subunits, integrin αv and integrin β5, that were expressed in beta cells. This finding was further elaborated using immunofluorescence analysis of histological sections derived from donor human pancreas. Despite the broad expression of specific integrin subunits, we found that expression of integrin αvβ5 heterodimers was restricted to beta cells and that this complex persisted in islet remnants of some type 1 diabetic individuals from which insulin expression had been lost. This study identifies αvβ5 heterodimers as a novel cell surface marker of human pancreatic beta cells, a finding that will aid in the identification and characterisation of this important cell type.
Collapse
Affiliation(s)
- Jacqueline V Schiesser
- Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, VIC, 3052, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Thomas Loudovaris
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia
| | - Helen E Thomas
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia.,Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, VIC, 3065, Australia
| | - Andrew G Elefanty
- Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, VIC, 3052, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, 3052, Australia.,Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Edouard G Stanley
- Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, VIC, 3052, Australia. .,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, 3052, Australia. .,Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia.
| |
Collapse
|
17
|
Proteome-wide and matrisome-specific alterations during human pancreas development and maturation. Nat Commun 2021; 12:1020. [PMID: 33589611 PMCID: PMC7884717 DOI: 10.1038/s41467-021-21261-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 01/19/2021] [Indexed: 01/31/2023] Open
Abstract
The extracellular matrix (ECM) is unique to each tissue and capable of guiding cell differentiation, migration, morphology, and function. The ECM proteome of different developmental stages has not been systematically studied in the human pancreas. In this study, we apply mass spectrometry-based quantitative proteomics strategies using N,N-dimethyl leucine isobaric tags to delineate proteome-wide and ECM-specific alterations in four age groups: fetal (18-20 weeks gestation), juvenile (5-16 years old), young adults (21-29 years old) and older adults (50-61 years old). We identify 3,523 proteins including 185 ECM proteins and quantify 117 of them. We detect previously unknown proteome and matrisome features during pancreas development and maturation. We also visualize specific ECM proteins of interest using immunofluorescent staining and investigate changes in ECM localization within islet or acinar compartments. This comprehensive proteomics analysis contributes to an improved understanding of the critical roles that ECM plays throughout human pancreas development and maturation.
Collapse
|
18
|
Cozzitorto C, Mueller L, Ruzittu S, Mah N, Willnow D, Darrigrand JF, Wilson H, Khosravinia D, Mahmoud AA, Risolino M, Selleri L, Spagnoli FM. A Specialized Niche in the Pancreatic Microenvironment Promotes Endocrine Differentiation. Dev Cell 2020; 55:150-162.e6. [PMID: 32857951 PMCID: PMC7720791 DOI: 10.1016/j.devcel.2020.08.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 05/11/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022]
Abstract
The interplay between pancreatic epithelium and the surrounding microenvironment is pivotal for pancreas formation and differentiation as well as adult organ homeostasis. The mesenchyme is the main component of the embryonic pancreatic microenvironment, yet its cellular identity is broadly defined, and whether it comprises functionally distinct cell subsets is not known. Using genetic lineage tracing, transcriptome, and functional studies, we identified mesenchymal populations with different roles during pancreatic development. Moreover, we showed that Pbx transcription factors act within the mouse pancreatic mesenchyme to define a pro-endocrine specialized niche. Pbx directs differentiation of endocrine progenitors into insulin- and glucagon-positive cells through non-cell-autonomous regulation of ECM-integrin interactions and soluble molecules. Next, we measured functional conservation between mouse and human pancreatic mesenchyme by testing identified mesenchymal factors in an iPSC-based differentiation model. Our findings provide insights into how lineage-specific crosstalk between epithelium and neighboring mesenchymal cells underpin the generation of different pancreatic cell types.
Collapse
Affiliation(s)
- Corinna Cozzitorto
- Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin 13125, Germany; Department of Ophthalmology & Department of Anatomy, Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Laura Mueller
- Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin 13125, Germany; Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Silvia Ruzittu
- Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin 13125, Germany; Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Nancy Mah
- Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - David Willnow
- Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin 13125, Germany; Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Jean-Francois Darrigrand
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Heather Wilson
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Daniel Khosravinia
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Amir-Ala Mahmoud
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Department of Orofacial Sciences & Department of Anatomy, University of California, San Francisco, 513 Parnassus Ave, HSW 710, San Francisco, CA 94143, USA
| | - Maurizio Risolino
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Department of Orofacial Sciences & Department of Anatomy, University of California, San Francisco, 513 Parnassus Ave, HSW 710, San Francisco, CA 94143, USA
| | - Licia Selleri
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Department of Orofacial Sciences & Department of Anatomy, University of California, San Francisco, 513 Parnassus Ave, HSW 710, San Francisco, CA 94143, USA
| | - Francesca M Spagnoli
- Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin 13125, Germany; Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK.
| |
Collapse
|
19
|
Massaro C, Sgueglia G, Frattolillo V, Baglio SR, Altucci L, Dell’Aversana C. Extracellular Vesicle-Based Nucleic Acid Delivery: Current Advances and Future Perspectives in Cancer Therapeutic Strategies. Pharmaceutics 2020; 12:pharmaceutics12100980. [PMID: 33081417 PMCID: PMC7589909 DOI: 10.3390/pharmaceutics12100980] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs) are sophisticated and sensitive messengers released by cells to communicate with and influence distant and neighboring cells via selective transfer of bioactive content, including protein lipids and nucleic acids. EVs have therefore attracted broad interest as new and refined potential therapeutic systems in many diseases, including cancer, due to their low immunogenicity, non-toxicity, and elevated bioavailability. They might serve as safe and effective vehicles for the transport of therapeutic molecules to specific tissues and cells. In this review, we focus on EVs as a vehicle for gene therapy in cancer. We describe recent developments in EV engineering to achieve efficient intracellular delivery of cancer therapeutics and avoid off-target effects, to provide an overview of the potential applications of EV-mediated gene therapy and the most promising biomedical advances.
Collapse
Affiliation(s)
- Crescenzo Massaro
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio 7, 80138 Naples, Italy; (C.M.); (G.S.); (V.F.)
| | - Giulia Sgueglia
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio 7, 80138 Naples, Italy; (C.M.); (G.S.); (V.F.)
| | - Victoria Frattolillo
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio 7, 80138 Naples, Italy; (C.M.); (G.S.); (V.F.)
| | - S. Rubina Baglio
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, de Boelelaan 1117, 1081HV Amsterdam, The Netherlands;
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio 7, 80138 Naples, Italy; (C.M.); (G.S.); (V.F.)
- Correspondence: (L.A.); (C.D.); Tel.: +39-081-5667569 (L.A.); +39-081-5667564 (C.D.)
| | - Carmela Dell’Aversana
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio 7, 80138 Naples, Italy; (C.M.); (G.S.); (V.F.)
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore” (IEOS)-National Research Council (CNR), Via Sergio Pansini 5, 80131 Naples, Italy
- Correspondence: (L.A.); (C.D.); Tel.: +39-081-5667569 (L.A.); +39-081-5667564 (C.D.)
| |
Collapse
|
20
|
Goh SK, Halfter W, Richardson T, Bertera S, Vaidya V, Candiello J, Bradford M, Banerjee I. Organ-specific ECM arrays for investigating Cell-ECM interactions during stem cell differentiation. Biofabrication 2020; 13. [PMID: 33045682 DOI: 10.1088/1758-5090/abc05f] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/12/2020] [Indexed: 12/22/2022]
Abstract
Pluripotent stem cells are promising source of cells for tissue engineering, regenerative medicine and drug discovery applications. The process of stem cell differentiation is regulated by multi-parametric cues from the surrounding microenvironment, one of the critical one being cell interaction with extracellular matrix (ECM). The ECM is a complex tissue-specific structure which are important physiological regulators of stem cell function and fate. Recapitulating this native ECM microenvironment niche is best facilitated by decellularized tissue/ organ derived ECM, which can faithfully reproduce the physiological environment with high fidelity to in vivo condition and promote tissue-specific cellular development and maturation. Recognizing the need for organ specific ECM in a 3D culture environment in driving phenotypic differentiation and maturation of hPSCs, we fabricated an ECM array platform using native-mimicry ECM from decellularized organs (namely pancreas, liver and heart), which allows cell-ECM interactions in both 2D and 3D configuration. The ECM array was integrated with rapid quantitative imaging for a systematic investigation of matrix protein profiles and sensitive measurement of cell-ECM interaction during hPSC differentiation. We tested our platform by elucidating the role of the three different organ-specific ECM in supporting induced pancreatic differentiation of hPSCs. While the focus of this report is on pancreatic differentiation, the developed platform is versatile to be applied to characterize any lineage specific differentiation.
Collapse
Affiliation(s)
- Saik Kia Goh
- University of Pittsburgh, Pittsburgh, 15261, UNITED STATES
| | - Willi Halfter
- University of Pittsburgh, Pittsburgh, Pennsylvania, UNITED STATES
| | - Thomas Richardson
- Chemical Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, UNITED STATES
| | - Suzanne Bertera
- Allegheny Health Network, Pittsburgh, Pennsylvania, UNITED STATES
| | - Vimal Vaidya
- University of Pittsburgh, Pittsburgh, Pennsylvania, UNITED STATES
| | - Joe Candiello
- University of Pittsburgh, Pittsburgh, Pennsylvania, UNITED STATES
| | - Mahalia Bradford
- Chemical Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, UNITED STATES
| | - Ipsita Banerjee
- Chemical Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, UNITED STATES
| |
Collapse
|
21
|
Almaça J, Caicedo A, Landsman L. Beta cell dysfunction in diabetes: the islet microenvironment as an unusual suspect. Diabetologia 2020; 63:2076-2085. [PMID: 32894318 PMCID: PMC7655222 DOI: 10.1007/s00125-020-05186-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022]
Abstract
Cells in different tissues, including endocrine cells in the pancreas, live in complex microenvironments that are rich in cellular and acellular components. Intricate interactions with their microenvironment dictate most cellular properties, such as their function, structure and size, and maintain tissue homeostasis. Pancreatic islets are populated by endocrine, vascular and immune cells that are immersed in the extracellular matrix. While the intrinsic properties of beta cells have been vastly investigated, our understanding of their interactions with their surroundings has only recently begun to unveil. Here, we review current research on the interplay between the islet cellular and acellular components, and the role these components play in beta cell physiology and pathophysiology. Although beta cell failure is a key pathomechanism in diabetes, its causes are far from being fully elucidated. We, thus, propose deleterious alterations of the islet niche as potential underlying mechanisms contributing to beta cell failure. In sum, this review emphasises that the function of the pancreatic islet depends on all of its components. Graphical abstract.
Collapse
Affiliation(s)
- Joana Almaça
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th avenue, Miami, FL, 33136, USA.
| | - Alejandro Caicedo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th avenue, Miami, FL, 33136, USA.
| | - Limor Landsman
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel.
| |
Collapse
|
22
|
Win PW, Oakie A, Li J, Wang R. Beta-cell β1 integrin deficiency affects in utero development of islet growth and vascularization. Cell Tissue Res 2020; 381:163-175. [PMID: 32060653 DOI: 10.1007/s00441-020-03179-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 01/27/2020] [Indexed: 10/25/2022]
Abstract
The β1 integrin subunit contributes to pancreatic beta cell growth and function through communication with the extracellular matrix (ECM). The effects of in vitro and in vivo β1 integrin knockout have been extensively studied in mature islets, yet no study to date has examined how the loss of β1 integrin during specific stages of pancreatic development impacts beta cell maturation. Beta-cell-specific tamoxifen-inducible Cre recombinase (MIP-CreERT) mice were crossed with mice containing floxed Itgb1 (β1 integrin) to create an inducible mouse model (MIPβ1KO) at the second transition stage (e13.5) of pancreas development. By e19.5-20.5, the expression of beta-cell β1 integrin in fetal MIPβ1KO mice was significantly reduced and these mice displayed decreased beta cell mass, density and proliferation. Morphologically, fetal MIPβ1KO pancreata exhibited reduced islet vascularization and nascent endocrine cells in the ductal region. In addition, decreased ERK phosphorylation was observed in fetal MIPβ1KO pancreata. The expression of transcription factors needed for beta-cell development was unchanged in fetal MIPβ1KO pancreata. The findings from this study demonstrate that β1 integrin signaling is required during a transition-specific window in the developing beta-cell to maintain islet mass and vascularization.
Collapse
Affiliation(s)
- Phyo Wei Win
- Children's Health Research Institute, Victoria Research Laboratories, London, Ontario, N6C 2V5, Canada.,Department of Physiology & Pharmacology, University of Western Ontario, London, Ontario, N6A 3K7, Canada
| | - Amanda Oakie
- Children's Health Research Institute, Victoria Research Laboratories, London, Ontario, N6C 2V5, Canada.,Department of Pathology & Laboratory Medicine, University of Western Ontario, London, Ontario, N6A 3K7, Canada
| | - Jinming Li
- Children's Health Research Institute, Victoria Research Laboratories, London, Ontario, N6C 2V5, Canada.,Department of Physiology & Pharmacology, University of Western Ontario, London, Ontario, N6A 3K7, Canada
| | - Rennian Wang
- Children's Health Research Institute, Victoria Research Laboratories, London, Ontario, N6C 2V5, Canada. .,Department of Physiology & Pharmacology, University of Western Ontario, London, Ontario, N6A 3K7, Canada.
| |
Collapse
|
23
|
Alessandra G, Algerta M, Paola M, Carsten S, Cristina L, Paolo M, Elisa M, Gabriella T, Carla P. Shaping Pancreatic β-Cell Differentiation and Functioning: The Influence of Mechanotransduction. Cells 2020; 9:E413. [PMID: 32053947 PMCID: PMC7072458 DOI: 10.3390/cells9020413] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/29/2020] [Accepted: 02/07/2020] [Indexed: 02/08/2023] Open
Abstract
Embryonic and pluripotent stem cells hold great promise in generating β-cells for both replacing medicine and novel therapeutic discoveries in diabetes mellitus. However, their differentiation in vitro is still inefficient, and functional studies reveal that most of these β-like cells still fail to fully mirror the adult β-cell physiology. For their proper growth and functioning, β-cells require a very specific environment, the islet niche, which provides a myriad of chemical and physical signals. While the nature and effects of chemical stimuli have been widely characterized, less is known about the mechanical signals. We here review the current status of knowledge of biophysical cues provided by the niche where β-cells normally live and differentiate, and we underline the possible machinery designated for mechanotransduction in β-cells. Although the regulatory mechanisms remain poorly understood, the analysis reveals that β-cells are equipped with all mechanosensors and signaling proteins actively involved in mechanotransduction in other cell types, and they respond to mechanical cues by changing their behavior. By engineering microenvironments mirroring the biophysical niche properties it is possible to elucidate the β-cell mechanotransductive-regulatory mechanisms and to harness them for the promotion of β-cell differentiation capacity in vitro.
Collapse
Affiliation(s)
- Galli Alessandra
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Marku Algerta
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Marciani Paola
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Schulte Carsten
- CIMAINA, Department of Physics, Università degli Studi di Milano, 20133 Milan, Italy
| | - Lenardi Cristina
- CIMAINA, Department of Physics, Università degli Studi di Milano, 20133 Milan, Italy
| | - Milani Paolo
- CIMAINA, Department of Physics, Università degli Studi di Milano, 20133 Milan, Italy
| | - Maffioli Elisa
- Department of Veterinary Medicine, Università degli Studi di Milano, 20133 Milan, Italy
| | - Tedeschi Gabriella
- Department of Veterinary Medicine, Università degli Studi di Milano, 20133 Milan, Italy
| | - Perego Carla
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| |
Collapse
|
24
|
Lammert E, Thorn P. The Role of the Islet Niche on Beta Cell Structure and Function. J Mol Biol 2019; 432:1407-1418. [PMID: 31711959 DOI: 10.1016/j.jmb.2019.10.032] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/25/2019] [Accepted: 10/29/2019] [Indexed: 01/15/2023]
Abstract
The islets of Langerhans or pancreatic islets are pivotal in the control of blood glucose and are complex microorgans embedded within the larger volume of the exocrine pancreas. Humans can have ~3.2 million islets [1] which, to our current knowledge, function in a similar manner to sense circulating blood glucose levels and respond with the secretion of a mix of different hormones that act to maintain glucose concentrations around a specific set point [2]. At a cellular level, the control of hormone secretion by glucose and other secretagogues is well-understood [3]. The key signal cascades have been identified and many details of the secretory process are known. However, if we shift focus from single cells and consider cells within intact islets, we do not have a comprehensive model as to how the islet environment influences cell function and how the islets work as a whole. This is important because there is overwhelming evidence that the structure and function of the individual endocrine cells are dramatically affected by the islet environment [4,5]. Uncovering the influence of this islet niche might drive future progress in treatments for Type 2 diabetes [6] and cell replacement therapies for Type 1 diabetes [7]. In this review, we focus on the insulin secreting beta cells and their interactions with the immediate environment that surrounds them including endocrine-endocrine interactions and contacts with capillaries.
Collapse
Affiliation(s)
- Eckhard Lammert
- Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany; Institute for Vascular and Islet Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Peter Thorn
- Charles Perkins Centre, School of Medical Sciences, University of Sydney, Camperdown, NSW 2006, Australia.
| |
Collapse
|
25
|
Townsend SE, Gannon M. Extracellular Matrix-Associated Factors Play Critical Roles in Regulating Pancreatic β-Cell Proliferation and Survival. Endocrinology 2019; 160:1885-1894. [PMID: 31271410 PMCID: PMC6656423 DOI: 10.1210/en.2019-00206] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/27/2019] [Indexed: 12/27/2022]
Abstract
This review describes formation of the islet basement membrane and the function of extracellular matrix (ECM) components in β-cell proliferation and survival. Implications for islet transplantation are discussed. The insulin-producing β-cell is key for maintaining glucose homeostasis. The islet microenvironment greatly influences β-cell survival and proliferation. Within the islet, β-cells contact the ECM, which is deposited primarily by intraislet endothelial cells, and this interaction has been shown to modulate proliferation and survival. ECM-localized growth factors, such as vascular endothelial growth factor and cellular communication network 2, signal through specific receptors and integrins on the β-cell surface. Further understanding of how the ECM functions to influence β-cell proliferation and survival will provide targets for enhancing functional β-cell mass for the treatment of diabetes.
Collapse
Affiliation(s)
- Shannon E Townsend
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
| | - Maureen Gannon
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
- Department of Veterans Affairs, Tennessee Valley Health Authority, Nashville, Tennessee
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Correspondence: Maureen Gannon, PhD, Vanderbilt University Medical Center, 2213 Garland Avenue, MRB IV 7465, Nashville, Tennessee 37232. E-mail:
| |
Collapse
|
26
|
Ma F, Tremmel DM, Li Z, Lietz CB, Sackett SD, Odorico JS, Li L. In Depth Quantification of Extracellular Matrix Proteins from Human Pancreas. J Proteome Res 2019; 18:3156-3165. [PMID: 31200599 DOI: 10.1021/acs.jproteome.9b00241] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Extracellular matrix (ECM) is an important component of the pancreatic microenvironment which regulates β cell proliferation, differentiation, and insulin secretion. Protocols have recently been developed for the decellularization of the human pancreas to generate functional scaffolds and hydrogels. In this work, we characterized human pancreatic ECM composition before and after decellularization using isobaric dimethylated leucine (DiLeu) labeling for relative quantification of ECM proteins. A novel correction factor was employed in the study to eliminate the bias introduced during sample preparation. In comparison to the commonly employed sample preparation methods (urea and FASP) for proteomic analysis, a recently developed surfactant and chaotropic agent assisted sequential extraction/on pellet digestion (SCAD) protocol has provided an improved strategy for ECM protein extraction of human pancreatic ECM matrix. The quantitative proteomic results revealed the preservation of matrisome proteins while most of the cellular proteins were removed. This method was compared with a well-established label-free quantification (LFQ) approach which rendered similar expressions of different categories of proteins (collagens, ECM glycoproteins, proteoglycans, etc.). The distinct expression of ECM proteins was quantified comparing adult and fetal pancreas ECM, shedding light on the correlation between matrix composition and postnatal β cell maturation. Despite the distinct profiles of different subcategories in the native pancreas, the distribution of matrisome proteins exhibited similar trends after the decellularization process. Our method generated a large data set of matrisome proteins from a single tissue type. These results provide valuable insight into the possibilities of constructing a bioengineered pancreas. It may also facilitate better understanding of the potential roles that matrisome proteins play in postnatal β cell maturation.
Collapse
Affiliation(s)
- Fengfei Ma
- School of Pharmacy , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Daniel M Tremmel
- Department of Surgery, Division of Transplantation, School of Medicine and Public Health , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Zihui Li
- Department of Chemistry , University of Wisconsin-Madison , Madison , Wisconsin 53706 , United States
| | - Christopher B Lietz
- Department of Chemistry , University of Wisconsin-Madison , Madison , Wisconsin 53706 , United States
| | - Sara Dutton Sackett
- Department of Surgery, Division of Transplantation, School of Medicine and Public Health , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Jon S Odorico
- Department of Surgery, Division of Transplantation, School of Medicine and Public Health , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Lingjun Li
- School of Pharmacy , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States.,Department of Chemistry , University of Wisconsin-Madison , Madison , Wisconsin 53706 , United States
| |
Collapse
|
27
|
Lee SN, Ahn JS, Lee SG, Lee HS, Choi AMK, Yoon JH. Integrins αvβ5 and αvβ6 Mediate IL-4–induced Collective Migration in Human Airway Epithelial Cells. Am J Respir Cell Mol Biol 2019; 60:420-433. [DOI: 10.1165/rcmb.2018-0081oc] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
| | | | - Seong Gyu Lee
- School of Mechanical Engineering, Yonsei University, Seoul, Korea
| | - Hyung-Suk Lee
- School of Mechanical Engineering, Yonsei University, Seoul, Korea
| | - Augustine M. K. Choi
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, New York; and
- Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, New York
| | - Joo-Heon Yoon
- The Airway Mucus Institute and
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
28
|
Harnessing CXCL12 signaling to protect and preserve functional β-cell mass and for cell replacement in type 1 diabetes. Pharmacol Ther 2019; 193:63-74. [DOI: 10.1016/j.pharmthera.2018.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
29
|
Pancreas organogenesis: The interplay between surrounding microenvironment(s) and epithelium-intrinsic factors. Curr Top Dev Biol 2019; 132:221-256. [DOI: 10.1016/bs.ctdb.2018.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
30
|
3D-Models of Insulin-Producing β-Cells: from Primary Islet Cells to Stem Cell-Derived Islets. Stem Cell Rev Rep 2018; 14:177-188. [PMID: 29181780 DOI: 10.1007/s12015-017-9783-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is a need for physiologically relevant assay platforms to provide functionally relevant models of diabetes, to accelerate the discovery of new treatment options and boost developments in drug discovery. In this review, we compare several 3D-strategies that have been used to increase the functional relevance of ex vivo human primary pancreatic islets and developments into the generation of stem cell derived pancreatic beta-cells (β-cells). Special attention will be given to recent approaches combining the use of extracellular matrix (ECM) scaffolds with pancreatic molecular memory, which can be used to improve yield and functionality of in vitro stem cell-derived pancreatic models. The ultimate goal is to develop scalable cell-based platforms for diabetes research and drug screening. This article will critically assess key aspects related to in vitro pancreatic 3D-ECM models and highlight the most promising approaches for future research.
Collapse
|
31
|
Olaniru OE, Persaud SJ. Identifying novel therapeutic targets for diabetes through improved understanding of islet adhesion receptors. Curr Opin Pharmacol 2018; 43:27-33. [DOI: 10.1016/j.coph.2018.07.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022]
|
32
|
Mamidi A, Prawiro C, Seymour PA, de Lichtenberg KH, Jackson A, Serup P, Semb H. Mechanosignalling via integrins directs fate decisions of pancreatic progenitors. Nature 2018; 564:114-118. [DOI: 10.1038/s41586-018-0762-2] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 10/19/2018] [Indexed: 12/18/2022]
|
33
|
Hadavi E, Leijten J, Brinkmann J, Jonkheijm P, Karperien M, van Apeldoorn A. Fibronectin and Collagen IV Microcontact Printing Improves Insulin Secretion by INS1E Cells. Tissue Eng Part C Methods 2018; 24:628-636. [PMID: 30306836 DOI: 10.1089/ten.tec.2018.0151] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
IMPACT STATEMENT This research deals with finding a proper bioengineering strategy for the creation of improved β-cell replacement therapy in type 1 diabetes. It specifically deals with the microenvironment of β-cells and its relationship to their endocrine function.
Collapse
Affiliation(s)
- Elahe Hadavi
- 1 Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente , Enschede, The Netherlands
| | - Jeroen Leijten
- 1 Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente , Enschede, The Netherlands
| | - Jenny Brinkmann
- 2 MESA+ Institute for Nanotechnology, Molecular Nanofabrication Group, University of Twente , Enschede, The Netherlands
| | - Pascal Jonkheijm
- 2 MESA+ Institute for Nanotechnology, Molecular Nanofabrication Group, University of Twente , Enschede, The Netherlands
| | - Marcel Karperien
- 1 Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente , Enschede, The Netherlands
| | - Aart van Apeldoorn
- 1 Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente , Enschede, The Netherlands .,3 Complex Tissue Regeneration Department, MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University , Maastricht, The Netherlands
| |
Collapse
|
34
|
Sackett SD, Tremmel DM, Ma F, Feeney AK, Maguire RM, Brown ME, Zhou Y, Li X, O'Brien C, Li L, Burlingham WJ, Odorico JS. Extracellular matrix scaffold and hydrogel derived from decellularized and delipidized human pancreas. Sci Rep 2018; 8:10452. [PMID: 29993013 PMCID: PMC6041318 DOI: 10.1038/s41598-018-28857-1] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/02/2018] [Indexed: 12/21/2022] Open
Abstract
Extracellular matrix (ECM) plays an important developmental role by regulating cell behaviour through structural and biochemical stimulation. Tissue-specific ECM, attained through decellularization, has been proposed in several strategies for tissue and organ replacement. Decellularization of animal pancreata has been reported, but the same methods applied to human pancreas are less effective due to higher lipid content. Moreover, ECM-derived hydrogels can be obtained from many decellularized tissues, but methods have not been reported to obtain human pancreas-derived hydrogel. Using novel decellularization methods with human pancreas we produced an acellular, 3D biological scaffold (hP-ECM) and hydrogel (hP-HG) amenable to tissue culture, transplantation and proteomic applications. The inclusion of a homogenization step in the decellularization protocol significantly improved lipid removal and gelation capability of the resulting ECM, which was capable of gelation at 37 °C in vitro and in vivo, and is cytocompatible with a variety of cell types and islet-like tissues in vitro. Overall, this study demonstrates the characterisation of a novel protocol for the decellularization and delipidization of human pancreatic tissue for the production of acellular ECM and ECM hydrogel suitable for cell culture and transplantation applications. We also report a list of 120 proteins present within the human pancreatic matrisome.
Collapse
Affiliation(s)
- Sara Dutton Sackett
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, 53705, USA.
| | - Daniel M Tremmel
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, 53705, USA
| | - Fengfei Ma
- School of Pharmacy, University of Wisconsin, Madison, Wisconsin, 53705, USA
| | - Austin K Feeney
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, 53705, USA
| | - Rachel M Maguire
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, 53705, USA
| | - Matthew E Brown
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, 53705, USA
| | - Ying Zhou
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, 53705, USA
| | - Xiang Li
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, 53705, USA
| | - Cori O'Brien
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, 53705, USA
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin, Madison, Wisconsin, 53705, USA
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, 53705, USA
| | - William J Burlingham
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, 53705, USA
| | - Jon S Odorico
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, 53705, USA
| |
Collapse
|
35
|
Llacua LA, Faas MM, de Vos P. Extracellular matrix molecules and their potential contribution to the function of transplanted pancreatic islets. Diabetologia 2018; 61:1261-1272. [PMID: 29306997 PMCID: PMC6449002 DOI: 10.1007/s00125-017-4524-8] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 10/18/2017] [Indexed: 12/18/2022]
Abstract
Extracellular matrix (ECM) molecules are responsible for structural and biochemical support, as well as for regulation of molecular signalling and tissue repair in many organ structures, including the pancreas. In pancreatic islets, collagen type IV and VI, and laminins are the most abundant molecules, but other ECM molecules are also present. The ECM interacts with specific combinations of integrin α/β heterodimers on islet cells and guides many cellular processes. More specifically, some ECM molecules are involved in beta cell survival, function and insulin production, while others can fine tune the susceptibility of islet cells to cytokines. Further, some ECM induce release of growth factors to facilitate tissue repair. During enzymatic isolation of islets for transplantation, the ECM is damaged, impacting islet function. However, restoration of the ECM in human islets (for example by adding ECM to the interior of immunoprotective capsules) has been shown to enhance islet function. Here, we provide current insight into the role of ECM molecules in islet function and discuss the clinical potential of ECM manipulation to enhance pancreatic islet function and survival.
Collapse
Affiliation(s)
- L Alberto Llacua
- Section of Immunoendocrinology, Department of Pathology and Medical Biology, University of Groningen, Hanzeplein 1 EA11, 9700 RB, Groningen, the Netherlands.
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Marijke M Faas
- Section of Immunoendocrinology, Department of Pathology and Medical Biology, University of Groningen, Hanzeplein 1 EA11, 9700 RB, Groningen, the Netherlands
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Paul de Vos
- Section of Immunoendocrinology, Department of Pathology and Medical Biology, University of Groningen, Hanzeplein 1 EA11, 9700 RB, Groningen, the Netherlands
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
36
|
Kim DS, Song L, Wang J, Wu H, Gu G, Sugi Y, Li Z, Wang H. GRP94 Is an Essential Regulator of Pancreatic β-Cell Development, Mass, and Function in Male Mice. Endocrinology 2018; 159:1062-1073. [PMID: 29272356 PMCID: PMC5793778 DOI: 10.1210/en.2017-00685] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 12/08/2017] [Indexed: 12/25/2022]
Abstract
Deficiencies in pancreatic β-cell mass contribute to both type 1 and type 2 diabetes. We investigated the role of the glucose-regulated protein (GRP) 94, an endoplasmic reticulum protein abundantly expressed in the pancreatic acini and islets, in β-cell development, survival, and function. We used a conditional knockout (KO) mouse in which the GRP94 gene, Hsp90b1, was specifically deleted in pancreatic and duodenal homeobox 1 (Pdx1)-expressing cells. These Hsp90b1 flox/flox;Pdx1Cre KO mice exhibited pancreatic hypoplasia at embryonic day (E) 16.5 to E18.5 and had significantly reduced β-cell mass at 4 weeks after birth. Further mechanistic studies showed that deletion of GRP94 reduced β-cell proliferation with increased cell apoptosis in both Pdx1+ endocrine progenitor cells and differentiated β cells. Although Hsp90b1 flox/flox;Pdx1Cre KO mice remained euglycemic at 8 weeks of age, they exhibited impaired glucose tolerance. In aggregate, these findings indicate that GRP94 is an essential regulator of pancreatic β-cell development, mass, and function.
Collapse
Affiliation(s)
- Do-sung Kim
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Lili Song
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Jingjing Wang
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Hongju Wu
- Department of Medicine, Tulane University, New Orleans, Louisiana 70112
| | - Guoqiang Gu
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37235
| | - Yukiko Sugi
- Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Zihai Li
- Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina 29425
| |
Collapse
|
37
|
Petersen MB, Gonçalves CA, Kim YH, Grapin-Botton A. Recapitulating and Deciphering Human Pancreas Development From Human Pluripotent Stem Cells in a Dish. Curr Top Dev Biol 2018; 129:143-190. [DOI: 10.1016/bs.ctdb.2018.02.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
38
|
Bastidas-Ponce A, Scheibner K, Lickert H, Bakhti M. Cellular and molecular mechanisms coordinating pancreas development. Development 2017; 144:2873-2888. [PMID: 28811309 DOI: 10.1242/dev.140756] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The pancreas is an endoderm-derived glandular organ that participates in the regulation of systemic glucose metabolism and food digestion through the function of its endocrine and exocrine compartments, respectively. While intensive research has explored the signaling pathways and transcriptional programs that govern pancreas development, much remains to be discovered regarding the cellular processes that orchestrate pancreas morphogenesis. Here, we discuss the developmental mechanisms and principles that are known to underlie pancreas development, from induction and lineage formation to morphogenesis and organogenesis. Elucidating such principles will help to identify novel candidate disease genes and unravel the pathogenesis of pancreas-related diseases, such as diabetes, pancreatitis and cancer.
Collapse
Affiliation(s)
- Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.,Technical University of Munich, Medical Faculty, 81675 Munich, Germany
| | - Katharina Scheibner
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.,Technical University of Munich, Medical Faculty, 81675 Munich, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.,Technical University of Munich, Medical Faculty, 81675 Munich, Germany
| | - Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany .,Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany
| |
Collapse
|
39
|
Llacua LA, de Haan BJ, de Vos P. Laminin and collagen IV inclusion in immunoisolating microcapsules reduces cytokine-mediated cell death in human pancreatic islets. J Tissue Eng Regen Med 2017; 12:460-467. [PMID: 28508555 DOI: 10.1002/term.2472] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 04/05/2017] [Accepted: 05/09/2017] [Indexed: 12/21/2022]
Abstract
Extracellular matrix (ECM) molecules have several functions in pancreatic islets, including provision of mechanical support and prevention of cytotoxicity during inflammation. During islet isolation, ECM connections are damaged, and are not restored after encapsulation and transplantation. Inclusion of specific combinations of collagen type IV and laminins in immunoisolating capsules can enhance survival of pancreatic islets. Here we investigated whether ECM can also enhance survival and lower susceptibility of human islets to cytokine-mediated cytotoxicity. To this end, human islets were encapsulated in alginate with collagen IV and either RGD, LRE or PDSGR, i.e. laminin sequences. Islets in capsules without ECM served as control. The encapsulated islets were exposed to IL-1β, IFN-γ and TNF-α for 24 and 72 h. All combinations of ECM improved the islet cell survival, and reduced necrosis and apoptosis after cytokine exposure (P < 0.01). Collagen IV-RGD and collagen IV-LRE reduced danger-associated molecular patterns (DAMPs) release from islets (P < 0.05). Moreover, collagen IV-RGD and collagen IV-PDSGR, but not collagen IV-LRE, reduced NO release from encapsulated human islets (P < 0.05). This reduction correlated with a higher oxygen consumption rate (OCR) of islets in capsules containing collagen IV-RGD and collagen IV-PDSGR. Islets in capsules with collagen IV-LRE showed more dysfunction, and OCR was not different from islets in control capsules without ECM. Our study demonstrates that incorporation of specific ECM molecules such as collagen type IV with the laminin sequences RGD and PDSGR in immunoisolated islets can protect against cytokine toxicity.
Collapse
Affiliation(s)
- L Alberto Llacua
- Immunoendocrinology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, RB, Groningen, The Netherlands
| | - Bart J de Haan
- Immunoendocrinology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, RB, Groningen, The Netherlands
| | - Paul de Vos
- Immunoendocrinology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, RB, Groningen, The Netherlands
| |
Collapse
|
40
|
Aamodt KI, Powers AC. Signals in the pancreatic islet microenvironment influence β-cell proliferation. Diabetes Obes Metab 2017; 19 Suppl 1:124-136. [PMID: 28880471 PMCID: PMC5679109 DOI: 10.1111/dom.13031] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 05/22/2017] [Accepted: 06/01/2017] [Indexed: 12/31/2022]
Abstract
The progressive loss of pancreatic β-cell mass that occurs in both type 1 and type 2 diabetes is a primary factor driving efforts to identify strategies for effectively increasing, enhancing or restoring β-cell mass. While factors that seem to influence β-cell proliferation in specific contexts have been described, reliable stimulation of human β-cell proliferation has remained a challenge. Importantly, β-cells exist in the context of a complex, integrated pancreatic islet microenvironment where they interact with other endocrine cells, vascular endothelial cells, extracellular matrix, neuronal projections and islet macrophages. This review highlights different components of the pancreatic microenvironment, and reviews what is known about how signaling that occurs between β-cells and these other components influences β-cell proliferation. Future efforts to further define the role of the pancreatic islet microenvironment on β-cell proliferation may lead to the development of successful approaches to increase or restore β-cell mass in diabetes.
Collapse
Affiliation(s)
- Kristie I. Aamodt
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alvin C. Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- VA Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
41
|
Atouf F, Choi Y, Fowler MJ, Poffenberger G, Vobecky J, Ta M, Chapman GB, Powers AC, Lumelsky NL. Generation of Islet-Like Hormone-Producing Cells In Vitro from Adult Human Pancreas. Cell Transplant 2017; 14:735-48. [PMID: 16454348 DOI: 10.3727/000000005783982602] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Transplantation of pancreatic islets can provide long-lasting insulin independence for diabetic patients, but the current islet supply is limited. Here we describe a new in vitro system that utilizes adult human pancreatic islet-enriched fractions to generate hormone-producing cells over 3–4 weeks of culture. By labeling proliferating cells with a retrovirus-expressing green fluorescent protein, we show that in this system hormone-producing cells are generated de novo. These hormone-producing cells aggregate to form islet-like cell clusters. The cell clusters, when tested in vitro, release insulin in response to glucose and other secretagogues. After transplantation into immunodeficient, nondiabetic mice, the islet-like cell clusters survive and release human insulin. We propose that this system will be useful as an experimental tool for investigating mechanisms for generating new islet cells from the postnatal pancreas, and for designing strategies to generate physiologically competent pancreatic islet cells ex vivo.
Collapse
Affiliation(s)
- Fouad Atouf
- Islet and Autoimmunity Branch, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-1453, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Liu JL, Kaddour N, Chowdhury S, Li Q, Gao ZH. Role of CCN5 (WNT1 inducible signaling pathway protein 2) in pancreatic islets. J Diabetes 2017; 9:462-474. [PMID: 27863006 DOI: 10.1111/1753-0407.12507] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/07/2016] [Indexed: 12/15/2022] Open
Abstract
In search of direct targets of insulin-like growth factor (IGF)-1 action, we discovered CCN5 (WNT1 inducible signaling pathway protein 2 [WISP2]) as a novel protein expressed in pancreatic β-cells. As a member of the "CCN" ( C ysteine-rich angiogenic inducer 61 [Cyr61], C onnective tissue growth factor [CTGF in humans], and N ephroblastoma overexpressed [Nov; in chickens]) family, the expression of CCN5/WISP2 is stimulated by IGF-1 together with Wnt signaling. When overexpressed in insulinoma cells, CCN5 promotes cell proliferation and cell survival against streptozotocin-induced cell death. The cell proliferation effect seems to be caused by AKT phosphorylation and increased cyclin D1 levels. These properties resemble those of CCN2/CTGF, another isoform of the CCN family, although CCN5 is the only one within the family of six proteins that lacks the C-terminal repeat. Treatment of primary mouse islets with recombinant CCN5 protein produced similar effects to those of gene transfection, indicating that either as a matricellular protein or a secreted growth factor, CCN5 stimulates β-cell proliferation and regeneration in a paracrine fashion. This review also discusses the regulation of CCN5/WISP2 by estrogen and its involvement in angiogenesis and tumorigenesis.
Collapse
Affiliation(s)
- Jun-Li Liu
- Fraser Laboratories, Department of Medicine, The Research Institute of McGill University Health Centre, Montreal, Canada
| | - Nancy Kaddour
- Fraser Laboratories, Department of Medicine, The Research Institute of McGill University Health Centre, Montreal, Canada
| | - Subrata Chowdhury
- Fraser Laboratories, Department of Medicine, The Research Institute of McGill University Health Centre, Montreal, Canada
| | - Qing Li
- Fraser Laboratories, Department of Medicine, The Research Institute of McGill University Health Centre, Montreal, Canada
| | - Zu-Hua Gao
- Department of Pathology, The Research Institute of McGill University Health Centre, Montreal, Canada
| |
Collapse
|
43
|
Arous C, Wehrle-Haller B. Role and impact of the extracellular matrix on integrin-mediated pancreatic β-cell functions. Biol Cell 2017; 109:223-237. [PMID: 28266044 DOI: 10.1111/boc.201600076] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 03/01/2017] [Accepted: 03/02/2017] [Indexed: 12/17/2022]
Abstract
Understanding the organisation and role of the extracellular matrix (ECM) in islets of Langerhans is critical for maintaining pancreatic β-cells, and to recognise and revert the physiopathology of diabetes. Indeed, integrin-mediated adhesion signalling in response to the pancreatic ECM plays crucial roles in β-cell survival and insulin secretion, two major functions, which are affected in diabetes. Here, we would like to present an update on the major components of the pancreatic ECM, their role during integrin-mediated cell-matrix adhesions and how they are affected during diabetes. To treat diabetes, a promising approach consists in replacing β-cells by transplantation. However, efficiency is low, because β-cells suffer of anoikis, due to enzymatic digestion of the pancreatic ECM, which affects the survival of insulin-secreting β-cells. The strategy of adding ECM components during transplantation, to reproduce the pancreatic microenvironment, is a challenging task, as many of the regulatory mechanisms that control ECM deposition and turnover are not sufficiently understood. A better comprehension of the impact of the ECM on the adhesion and integrin-dependent signalling in β-cells is primordial to improve the healthy state of islets to prevent the onset of diabetes as well as for enhancing the efficiency of the islet transplantation therapy.
Collapse
Affiliation(s)
- Caroline Arous
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
| | - Bernhard Wehrle-Haller
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
| |
Collapse
|
44
|
Synergism of highly transducible adenovirus encoding heme oxygenase 1 gene and low-dose immunosuppressants for successful outcomes of xenotransplanted pancreatic islet. J IND ENG CHEM 2017. [DOI: 10.1016/j.jiec.2016.11.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
45
|
Cross SE, Vaughan RH, Willcox AJ, McBride AJ, Abraham AA, Han B, Johnson JD, Maillard E, Bateman PA, Ramracheya RD, Rorsman P, Kadler KE, Dunne MJ, Hughes SJ, Johnson PRV. Key Matrix Proteins Within the Pancreatic Islet Basement Membrane Are Differentially Digested During Human Islet Isolation. Am J Transplant 2017; 17:451-461. [PMID: 27456745 DOI: 10.1111/ajt.13975] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 07/12/2016] [Indexed: 02/06/2023]
Abstract
Clinical islet transplantation achieves insulin independence in selected patients, yet current methods for extracting islets from their surrounding pancreatic matrix are suboptimal. The islet basement membrane (BM) influences islet function and survival and is a critical marker of islet integrity following rodent islet isolation. No studies have investigated the impact of islet isolation on BM integrity in human islets, which have a unique duplex structure. To address this, samples were taken from 27 clinical human islet isolations (donor age 41-59, BMI 26-38, cold ischemic time < 10 h). Collagen IV, pan-laminin, perlecan and laminin-α5 in the islet BM were significantly digested by enzyme treatment. In isolated islets, laminin-α5 (found in both layers of the duplex BM) and perlecan were lost entirely, with no restoration evident during culture. Collagen IV and pan-laminin were present in the disorganized BM of isolated islets, yet a significant reduction in pan-laminin was seen during the initial 24 h culture period. Islet cytotoxicity increased during culture. Therefore, the human islet BM is substantially disrupted during the islet isolation procedure. Islet function and survival may be compromised as a consequence of an incomplete islet BM, which has implications for islet survival and transplanted graft longevity.
Collapse
Affiliation(s)
- S E Cross
- Islet Transplant Research Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, University of Oxford, Oxford, UK
| | - R H Vaughan
- Islet Transplant Research Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, University of Oxford, Oxford, UK
| | - A J Willcox
- Islet Transplant Research Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, University of Oxford, Oxford, UK
| | - A J McBride
- Islet Transplant Research Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, University of Oxford, Oxford, UK
| | - A A Abraham
- Islet Transplant Research Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, University of Oxford, Oxford, UK
| | - B Han
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - J D Johnson
- Islet Transplant Research Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, University of Oxford, Oxford, UK
| | - E Maillard
- Islet Transplant Research Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, University of Oxford, Oxford, UK
| | - P A Bateman
- Islet Transplant Research Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, University of Oxford, Oxford, UK
| | - R D Ramracheya
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, University of Oxford, Oxford, UK
| | - P Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, University of Oxford, Oxford, UK
| | - K E Kadler
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - M J Dunne
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - S J Hughes
- Islet Transplant Research Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, University of Oxford, Oxford, UK
| | - P R V Johnson
- Islet Transplant Research Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, University of Oxford, Oxford, UK
| |
Collapse
|
46
|
Larsen HL, Grapin-Botton A. The molecular and morphogenetic basis of pancreas organogenesis. Semin Cell Dev Biol 2017; 66:51-68. [PMID: 28089869 DOI: 10.1016/j.semcdb.2017.01.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 01/08/2023]
Abstract
The pancreas is an essential endoderm-derived organ that ensures nutrient metabolism via its endocrine and exocrine functions. Here we review the essential processes governing the embryonic and early postnatal development of the pancreas discussing both the mechanisms and molecules controlling progenitor specification, expansion and differentiation. We elaborate on how these processes are orchestrated in space and coordinated with morphogenesis. We draw mainly from experiments conducted in the mouse model but also from investigations in other model organisms, complementing a recent comprehensive review of human pancreas development (Jennings et al., 2015) [1]. The understanding of pancreas development in model organisms provides a framework to interpret how human mutations lead to neonatal diabetes and may contribute to other forms of diabetes and to guide the production of desired pancreatic cell types from pluripotent stem cells for therapeutic purposes.
Collapse
Affiliation(s)
- Hjalte List Larsen
- DanStem, University of Copenhagen, 3 B Blegdamsvej, DK-2200 Copenhagen N, Denmark
| | - Anne Grapin-Botton
- DanStem, University of Copenhagen, 3 B Blegdamsvej, DK-2200 Copenhagen N, Denmark.
| |
Collapse
|
47
|
Kaviani M, Azarpira N. Insight into microenvironment remodeling in pancreatic endocrine tissue engineering: Biological and biomaterial approaches. Tissue Eng Regen Med 2016; 13:475-484. [PMID: 30603429 PMCID: PMC6170842 DOI: 10.1007/s13770-016-0014-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 02/03/2016] [Accepted: 02/05/2016] [Indexed: 01/04/2023] Open
Abstract
The treatment of diabetes mellitus, as a chronic and complicated disease, is a valuable purpose. Islet transplantation can provide metabolic stability and insulin independence in type 1 diabetes patients. Diet and insulin therapy are only diabetes controllers and cannot remove all of the diabetes complications. Moreover, islet transplantation is more promising treatment than whole pancreas transplantation because of lesser invasive surgical procedure and morbidity and mortality. According to the importance of extracellular matrix for islet viability and function, microenvironment remodeling of pancreatic endocrine tissue can lead to more success in diabetes treatment by pancreatic islets. Production of bioengineered pancreas and remodeling of pancreas extracellular matrix provide essential microenvironment for re-vascularization, re-innervation and signaling cascades triggering. Therefore, islets show better viability and function in these conditions. Researchers conduct various scaffolds with different biomaterials for the improvement of islet viability, function and transplantation outcome. The attention to normal pancreas anatomy, embryology and histology is critical to understand the pancreatic Langerhans islets niche and finally to achieve efficient engineered structure. Therefore, in the present study, the status and components of the islets niche is mentioned and fundamental issues related to the tissue engineering of this structure is considered. The purpose of this review article is summarization of recent progress in the endocrine pancreas tissue engineering and biomaterials and biological aspects of it.
Collapse
Affiliation(s)
- Maryam Kaviani
- Transplant Research Center, Shiraz University of Medical Sciences, Mohamad Rasulallah Research Tower, Khalili street, Shiraz, 7193635899 Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Mohamad Rasulallah Research Tower, Khalili street, Shiraz, 7193635899 Iran
| |
Collapse
|
48
|
Pham H, Birtolo C, Chheda C, Yang W, Rodriguez MD, Liu ST, Gugliotta G, Lewis MS, Cirulli V, Pandol SJ, Ptasznik A. Essential Role of Lyn in Fibrosis. Front Physiol 2016; 7:387. [PMID: 27630579 PMCID: PMC5006658 DOI: 10.3389/fphys.2016.00387] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 08/22/2016] [Indexed: 12/13/2022] Open
Abstract
Fibrotic disorders involve replacement of normal parenchyma with myofibroblasts, which deposit connective tissue, leading to obliteration of the function of the underlying organ. The treatment options are inadequate and reflect the fact that signaling targets in myofibroblasts are unknown. Here we identify the hyperactive Lyn signaling in myofibroblasts of patients with chronic pancreatitis-induced fibrosis. Lyn activation coexpress with markers of activated myofibroblasts, and is increased ~11-fold in chronic pancreatitis compared to normal tissue. Inhibition of Lyn with siRNA or INNO-406 leads to the substantial decrease of migration and proliferation of human chronic pancreatitis myofibroblasts in vitro, while leaving migration and proliferation of normal myofibroblasts only slightly affected. Furthermore, inhibition of Lyn prevents synthesis of procollagen and collagen in myofibroblasts in a mouse model of chronic pancreatitis-induced fibrosis. We conclude that Lyn, as a positive regulator of myofibroblast migration, proliferation, and collagen production, is a key target for preventing fibrosis.
Collapse
Affiliation(s)
- Hung Pham
- Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical CenterLos Angeles, CA, USA; Department of Veterans AffairsLos Angeles, CA, USA; Department of Medicine, University of California, Los AngelesLos Angeles, CA, USA
| | - Chiara Birtolo
- Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical CenterLos Angeles, CA, USA; Department of Internal Medicine, University of BolognaBologna, Italy
| | - Chintan Chheda
- Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center Los Angeles, CA, USA
| | - Wendy Yang
- Department of Medicine, Institute of Stem Cell and Regenerative Medicine, University of Washington Seattle, WA, USA
| | - Maria D Rodriguez
- Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center Los Angeles, CA, USA
| | - Sandy T Liu
- Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical CenterLos Angeles, CA, USA; Department of Medicine, University of California, Los AngelesLos Angeles, CA, USA
| | - Gabriele Gugliotta
- Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical CenterLos Angeles, CA, USA; Department of Internal Medicine, University of BolognaBologna, Italy
| | - Michael S Lewis
- Department of Veterans AffairsLos Angeles, CA, USA; Department of Medicine, University of California, Los AngelesLos Angeles, CA, USA
| | - Vincenzo Cirulli
- Department of Medicine, Institute of Stem Cell and Regenerative Medicine, University of Washington Seattle, WA, USA
| | - Stephen J Pandol
- Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical CenterLos Angeles, CA, USA; Department of Veterans AffairsLos Angeles, CA, USA; Department of Medicine, University of California, Los AngelesLos Angeles, CA, USA
| | - Andrzej Ptasznik
- Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center Los Angeles, CA, USA
| |
Collapse
|
49
|
Henley KD, Stanescu DE, Kropp PA, Wright CVE, Won KJ, Stoffers DA, Gannon M. Threshold-Dependent Cooperativity of Pdx1 and Oc1 in Pancreatic Progenitors Establishes Competency for Endocrine Differentiation and β-Cell Function. Cell Rep 2016; 15:2637-2650. [PMID: 27292642 DOI: 10.1016/j.celrep.2016.05.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 02/26/2016] [Accepted: 05/09/2016] [Indexed: 02/06/2023] Open
Abstract
Pdx1 and Oc1 are co-expressed in multipotent pancreatic progenitors and regulate the pro-endocrine gene Neurog3. Their expression diverges in later organogenesis, with Oc1 absent from hormone+ cells and Pdx1 maintained in mature β cells. In a classical genetic test for cooperative functional interactions, we derived mice with combined Pdx1 and Oc1 heterozygosity. Endocrine development in double-heterozygous pancreata was normal at embryonic day (E)13.5, but defects in specification and differentiation were apparent at E15.5, the height of the second wave of differentiation. Pancreata from double heterozygotes showed alterations in the expression of genes crucial for β-cell development and function, decreased numbers and altered allocation of Neurog3-expressing endocrine progenitors, and defective endocrine differentiation. Defects in islet gene expression and β-cell function persisted in double heterozygous neonates. These results suggest that Oc1 and Pdx1 cooperate prior to their divergence, in pancreatic progenitors, to allow for proper differentiation and functional maturation of β cells.
Collapse
Affiliation(s)
- Kathryn D Henley
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232.,Program in Developmental Biology, Vanderbilt University, Nashville, TN 37232
| | - Diana E Stanescu
- Institute for Diabetes, Obesity and Metabolism and the Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104.,Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia PA 19104
| | - Peter A Kropp
- Program in Developmental Biology, Vanderbilt University, Nashville, TN 37232.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232
| | - Christopher V E Wright
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232.,Program in Developmental Biology, Vanderbilt University, Nashville, TN 37232
| | - Kyoung-Jae Won
- Institute for Diabetes, Obesity and Metabolism and the Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Doris A Stoffers
- Institute for Diabetes, Obesity and Metabolism and the Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Maureen Gannon
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232.,Program in Developmental Biology, Vanderbilt University, Nashville, TN 37232.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232.,Department of Medicine, Vanderbilt University, Nashville, TN 37232.,Department of Veterans Affairs, Tennessee Valley Health Authority, Vanderbilt University, Nashville, TN 37212
| |
Collapse
|
50
|
van de Bunt M, Lako M, Barrett A, Gloyn AL, Hansson M, McCarthy MI, Beer NL, Honoré C. Insights into islet development and biology through characterization of a human iPSC-derived endocrine pancreas model. Islets 2016; 8:83-95. [PMID: 27246810 PMCID: PMC4987020 DOI: 10.1080/19382014.2016.1182276] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Directed differentiation of stem cells offers a scalable solution to the need for human cell models recapitulating islet biology and T2D pathogenesis. We profiled mRNA expression at 6 stages of an induced pluripotent stem cell (iPSC) model of endocrine pancreas development from 2 donors, and characterized the distinct transcriptomic profiles associated with each stage. Established regulators of endodermal lineage commitment, such as SOX17 (log2 fold change [FC] compared to iPSCs = 14.2, p-value = 4.9 × 10(-5)) and the pancreatic agenesis gene GATA6 (log2 FC = 12.1, p-value = 8.6 × 10(-5)), showed transcriptional variation consistent with their known developmental roles. However, these analyses highlighted many other genes with stage-specific expression patterns, some of which may be novel drivers or markers of islet development. For example, the leptin receptor gene, LEPR, was most highly expressed in published data from in vivo-matured cells compared to our endocrine pancreas-like cells (log2 FC = 5.5, p-value = 2.0 × 10(-12)), suggesting a role for the leptin pathway in the maturation process. Endocrine pancreas-like cells showed significant stage-selective expression of adult islet genes, including INS, ABCC8, and GLP1R, and enrichment of relevant GO-terms (e.g. "insulin secretion"; odds ratio = 4.2, p-value = 1.9 × 10(-3)): however, principal component analysis indicated that in vitro-differentiated cells were more immature than adult islets. Integration of the stage-specific expression information with genetic data from T2D genome-wide association studies revealed that 46 of 82 T2D-associated loci harbor genes present in at least one developmental stage, facilitating refinement of potential effector transcripts. Together, these data show that expression profiling in an iPSC islet development model can further understanding of islet biology and T2D pathogenesis.
Collapse
Affiliation(s)
- Martijn van de Bunt
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Majlinda Lako
- Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom
| | - Amy Barrett
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Anna L. Gloyn
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
- Oxford NIHR Biomedical Research Center, Churchill Hospital, Oxford, United Kingdom
| | - Mattias Hansson
- Department of Diabetes Research, Novo Nordisk A/S, Maaloev, Denmark
| | - Mark I. McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
- Oxford NIHR Biomedical Research Center, Churchill Hospital, Oxford, United Kingdom
| | - Nicola L. Beer
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
- CONTACT Dr Nicola L Beer Oxford Center for Diabetes Endocrinology & Metabolism, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Christian Honoré
- Department of Islet and Stem Cell Biology, Novo Nordisk A/S, Maaloev, Denmark
| |
Collapse
|