1
|
Alver CG, Dominguez-Bendala J, Agarwal A. Engineered tools to study endocrine dysfunction of pancreas. BIOPHYSICS REVIEWS 2024; 5:041303. [PMID: 39449867 PMCID: PMC11498943 DOI: 10.1063/5.0220396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/04/2024] [Indexed: 10/26/2024]
Abstract
Pancreas, a vital organ with intricate endocrine and exocrine functions, is central to the regulation of the body's glucose levels and digestive processes. Disruptions in its endocrine functions, primarily regulated by islets of Langerhans, can lead to debilitating diseases such as diabetes mellitus. Murine models of pancreatic dysfunction have contributed significantly to the understanding of insulitis, islet-relevant immunological responses, and the optimization of cell therapies. However, genetic differences between mice and humans have severely limited their clinical translational relevance. Recent advancements in tissue engineering and microfabrication have ushered in a new era of in vitro models that offer a promising solution. This paper reviews the state-of-the-art engineered tools designed to study endocrine dysfunction of the pancreas. Islet on a chip devices that allow precise control of various culture conditions and noninvasive readouts of functional outcomes have led to the generation of physiomimetic niches for primary and stem cell derived islets. Live pancreatic slices are a new experimental tool that could more comprehensively recapitulate the complex cellular interplay between the endocrine and exocrine parts of the pancreas. Although a powerful tool, live pancreatic slices require more complex control over their culture parameters such as local oxygenation and continuous removal of digestive enzymes and cellular waste products for maintaining experimental functionality over long term. The combination of islet-immune and slice on chip strategies can guide the path toward the next generation of pancreatic tissue modeling for better understanding and treatment of endocrine pancreatic dysfunctions.
Collapse
Affiliation(s)
| | - Juan Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Ashutosh Agarwal
- Author to whom correspondence should be addressed:. Tel.: +1 305 243-8925
| |
Collapse
|
2
|
Avgoustiniatos ES, Mueller KR, Scott III WE, Kitzmann JP, Suszynski TM, Perrault BE, Falde EJ, Balamurugan AN, Hering BJ, Putnam CW, Papas KK. Silicone rubber membrane devices permit islet culture at high density without adverse effects. Front Bioeng Biotechnol 2024; 12:1401608. [PMID: 39070161 PMCID: PMC11273363 DOI: 10.3389/fbioe.2024.1401608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/21/2024] [Indexed: 07/30/2024] Open
Abstract
Introduction Conventional culture conditions, such as in T-flasks, require that oxygen diffuse through the medium to reach the islets; in turn, islet surface area density is limited by oxygen availability. To culture a typical clinical islet preparation may require more than 20 T-175 flasks at the standard surface area density of 200 IE/cm2. To circumvent this logistical constraint, we tested islets cultured on top of silicon gas-permeable (GP) membranes which place islets in close proximity to ambient oxygen. Methods Oxygenation of individual islets under three culture conditions, standard low-density, non-GP high density, and GP high density, were first modeled with finite element simulations. Porcine islets from 30 preparations were cultured for 2 days in devices with GP membrane bottoms or in paired cultures under conventional conditions. Islets were seeded at high density (HD, ∼4000 IE/cm2, as measured by DNA) in both GP and non-GP devices. Results In simulations, individual islets under standard culture conditions and high density cultures on GP membranes were both well oxygenated whereas non-GP high density cultured islets were anoxic. Similarly, compared to the non-GP paired controls, islet viability and recovery were significantly increased in HD GP cultures. The diabetes reversal rate in nude diabetic mice was similar for HD GP devices and standard cultures but was minimal with non-GP HD cultures. Discussion Culturing islets in GP devices allows for a 20-fold increase of islet surface area density, greatly simplifying the culture process while maintaining islet viability and metabolism.
Collapse
Affiliation(s)
| | - Kate R. Mueller
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, United States
| | - William E. Scott III
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, United States
| | - Jennifer P. Kitzmann
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, United States
- Department of Surgery, Institute for Cellular Transplantation, University of Arizona, Tucson, AZ, United States
| | - Thomas M. Suszynski
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, United States
| | - Brian E. Perrault
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, United States
| | - Eric J. Falde
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, United States
| | - A. N. Balamurugan
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, United States
| | - Bernhard J. Hering
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, United States
| | - Charles W. Putnam
- Department of Surgery, Institute for Cellular Transplantation, University of Arizona, Tucson, AZ, United States
| | - Klearchos K. Papas
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, United States
- Department of Surgery, Institute for Cellular Transplantation, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
3
|
Alver CG, Álvarez-Cubela S, Altilio I, Hutchison E, Warrner E, Viso ME, Vitale G, Oliver D, Pastori RL, Dominguez-Bendala J, Agarwal A. SliceChip: a benchtop fluidic platform for organotypic culture and serial assessment of human and rodent pancreatic slices. LAB ON A CHIP 2024; 24:1557-1572. [PMID: 38205530 PMCID: PMC10939771 DOI: 10.1039/d3lc00850a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 01/06/2024] [Indexed: 01/12/2024]
Abstract
Enzymatically isolated pancreatic islets are the most commonly used ex vivo testbeds for diabetes research. Recently, precision-cut living slices of human pancreas are emerging as an exciting alternative because they maintain the complex architecture of the endocrine and exocrine tissues, and do not suffer from the mechanical and chemical stress of enzymatic isolation. We report a fluidic pancreatic SliceChip platform with dynamic environmental controls that generates a warm, oxygenated, and bubble-free fluidic pathway across singular immobilized slices with continuous deliver of fresh media and the ability to perform repeat serial perfusion assessments. A degasser ensures the system remains bubble-free while systemic pressurization with compressed oxygen ensures slice medium remains adequately oxygenated. Computational modeling of perfusion and oxygen dynamics within SliceChip guide the system's physiomimetic culture conditions. Maintenance of the physiological glucose dependent insulin secretion profile across repeat perfusion assessments of individual pancreatic slices kept under physiological oxygen levels demonstrated the culture capacity of our platform. Fluorescent images acquired every 4 hours of transgenic murine pancreatic slices were reliably stable and recoverable over a 5 day period due to the inclusion of a 3D-printed bioinert metallic anchor that maintained slice position within the SliceChip. Our slice on a chip platform has the potential to expand the useability of human pancreatic slices for diabetes pathogenesis and the development of new therapeutic approaches, while also enabling organotypic culture and assessment of other tissue slices such as brain and patient tumors.
Collapse
Affiliation(s)
- Charles G Alver
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
- Medical Scientist Training Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Silvia Álvarez-Cubela
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Isabella Altilio
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Emily Hutchison
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - Emma Warrner
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - Mariana E Viso
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - Giana Vitale
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - David Oliver
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - Ricardo L Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Juan Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
4
|
Huang H, Karanth SS, Guan Y, Freeman S, Soron R, Godovich DS, Guan J, Ye K, Jin S. Oxygenated Scaffolds for Pancreatic Endocrine Differentiation from Induced Pluripotent Stem Cells. Adv Healthc Mater 2024; 13:e2302275. [PMID: 37885129 PMCID: PMC11578060 DOI: 10.1002/adhm.202302275] [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: 07/17/2023] [Revised: 10/06/2023] [Indexed: 10/28/2023]
Abstract
A 3D microenvironment is known to endorse pancreatic islet development from human induced pluripotent stem cells (iPSCs). However, oxygen supply becomes a limiting factor in a scaffold culture. In this study, oxygen-releasing biomaterials are fabricated and an oxygenated scaffold culture platform is developed to offer a better oxygen supply during 3D iPSC pancreatic differentiation. It is found that the oxygenation does not alter the scaffold's mechanical properties. The in situ oxygenation improves oxygen tension within the scaffolds. The unique 3D differentiation system enables the generation of islet organoids with enhanced expression of islet signature genes and proteins. Additionally, it is discovered that the oxygenation at the early stage of differentiation has more profound impacts on islet development from iPSCs. More C-peptide+ /MAFA+ β and glucagon+ /MAFB+ α cells formed in the iPSC-derived islet organoids generated under oxygenated conditions, suggesting enhanced maturation of the organoids. Furthermore, the oxygenated 3D cultures improve islet organoids' sensitivity to glucose for insulin secretion. It is herein demonstrated that the oxygenated scaffold culture empowers iPSC islet differentiation to generate clinically relevant tissues for diabetes research and treatment.
Collapse
Affiliation(s)
- Hui Huang
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Sciences, State University of New York (SUNY) at Binghamton, New York, 13902, USA
| | - Soujanya S Karanth
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Sciences, State University of New York (SUNY) at Binghamton, New York, 13902, USA
| | - Ya Guan
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Sebastian Freeman
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Sciences, State University of New York (SUNY) at Binghamton, New York, 13902, USA
| | - Ryan Soron
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Sciences, State University of New York (SUNY) at Binghamton, New York, 13902, USA
| | - David S Godovich
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Sciences, State University of New York (SUNY) at Binghamton, New York, 13902, USA
| | - Jianjun Guan
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Kaiming Ye
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Sciences, State University of New York (SUNY) at Binghamton, New York, 13902, USA
- Center of Biomanufacturing for Regenerative Medicine, State University of New York (SUNY) at Binghamton, New York, 13902, USA
| | - Sha Jin
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Sciences, State University of New York (SUNY) at Binghamton, New York, 13902, USA
- Center of Biomanufacturing for Regenerative Medicine, State University of New York (SUNY) at Binghamton, New York, 13902, USA
| |
Collapse
|
5
|
Zheng H, Yin Z, Luo X, Zhou Y, Zhang F, Guo Z. Association of per- and polyfluoroalkyl substance exposure with metabolic syndrome and its components in adults and adolescents. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:112943-112958. [PMID: 37845597 PMCID: PMC10643431 DOI: 10.1007/s11356-023-30317-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 10/03/2023] [Indexed: 10/18/2023]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are widespread contaminants, but few studies have explored the relationship between PFAS and levels of metabolic syndrome (MetS) in the population. The available evidence of an association is also conflicting. We selected adults and adolescents with complete PFAS data from the National Health and Nutrition Examination Survey conducted between 2003 and 2018. We analyzed the association between PFAS and MetS using multivariate logistic regression models and evaluated potential nonlinear relationships with restricted cubic spline models. Additionally, we employed weighted quantile sum (WQS) regressions to uncover the multiple exposure effects and relative weights of each PFAS. Finally, we conducted a series of sensitivity analyses to test the robustness of our findings. In this population-based study, we analyzed data from a total of 4,973 adults, aged 20-85 years, and 1,381 adolescents, aged 12-19 years. Using fully adjusted multivariate logistic regression models, we found that serum levels of perfluorodecanoate (PFDA) [0.65 (0.50, 0.85)] and total PFAS [0.92 (0.85, 0.99)] were negatively associated with the prevalence of MetS in adults. Similarly, in adolescents, we observed negative correlations between the prevalence of MetS and levels of PFDA [0.55 (0.38, 0.80)], perfluorooctanoic acid (PFOA) [0.62 (0.39, 1.00)], perfluorooctane sulfonic acid (PFOS) [0.59 (0.36, 0.96)], and total PFAS [0.61 (0.37, 0.99)]. Additionally, our study identified statistically significant negative associations between serum levels of PFAS and certain components of MetS, primarily elevated fasting glucose and lower high-density lipoprotein cholesterol. Our study found that PFAS was associated with a lower prevalence of MetS in both adults and adolescents, offering new insights into the relationship between PFAS and metabolic health. Interestingly, however, we observed conflicting findings across the components of MetS. Specifically, we observed that PFAS had a negative correlation with some metrics and a positive correlation with others. These conflicting results point to a complex interplay between PFAS and various metrics of metabolic health.
Collapse
Affiliation(s)
- Huizhen Zheng
- Department of Cardiology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ziwei Yin
- Department of Cardiology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Xi Luo
- Department of Cardiology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yingli Zhou
- Department of Cardiology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Fei Zhang
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zhihua Guo
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China.
- Hunan Key Laboratory of Colleges of Intelligent Traditional Chinese Medicine Diagnosis and Preventive Treatment of Chronic Diseases, Changsha, 410208, China.
| |
Collapse
|
6
|
Yoshihara E. Adapting Physiology in Functional Human Islet Organogenesis. Front Cell Dev Biol 2022; 10:854604. [PMID: 35557947 PMCID: PMC9086403 DOI: 10.3389/fcell.2022.854604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/22/2022] [Indexed: 01/07/2023] Open
Abstract
Generation of three-dimensional (3D)-structured functional human islets is expected to be an alternative cell source for cadaveric human islet transplantation for the treatment of insulin-dependent diabetes. Human pluripotent stem cells (hPSCs), such as human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), offer infinite resources for newly synthesized human islets. Recent advancements in hPSCs technology have enabled direct differentiation to human islet-like clusters, which can sense glucose and secrete insulin, and those islet clusters can ameliorate diabetes when transplanted into rodents or non-human primates (NHPs). However, the generated hPSC-derived human islet-like clusters are functionally immature compared with primary human islets. There remains a challenge to establish a technology to create fully functional human islets in vitro, which are functionally and transcriptionally indistinguishable from cadaveric human islets. Understanding the complex differentiation and maturation pathway is necessary to generate fully functional human islets for a tremendous supply of high-quality human islets with less batch-to-batch difference for millions of patients. In this review, I summarized the current progress in the generation of 3D-structured human islets from pluripotent stem cells and discussed the importance of adapting physiology for in vitro functional human islet organogenesis and possible improvements with environmental cues.
Collapse
Affiliation(s)
- Eiji Yoshihara
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| |
Collapse
|
7
|
Optical sensor arrays designed for guided manufacture of perfluorocarbon nanoemulsions with a non-synthetic stabilizer. Acta Biomater 2021; 136:558-569. [PMID: 34563723 DOI: 10.1016/j.actbio.2021.09.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 02/08/2023]
Abstract
Hydrophobic drugs are incorporated into oil-in-water nanoemulsions (OIW) either as new formulations or repurposed for intravenous delivery. Typically, these are manufactured through stepwise processes of sonication or high-pressure homogenization (HPH). The guiding criteria for most nanoemulsion manufacture are the size and homogeneity/polydispersity of the drug-laden particles with strict requirements for clinical injectables. To date, most formulation optimization is done through trial and error with stepwise sampling during processing utilizing dynamic light scattering (DLS), light obscuration sensing (LOS) or laser particle tracking (LPT) to assess manufacturing progress. The objective of this work was to develop and implement an in-line optical turbidity/nephelometry sensor array for the longitudinal in-process monitoring of nanoemulsion manufacture. A further objective was the use of this sensor array to rapidly optimize the manufacture of a sub-120 nm oxygen carrying perfluorocarbon nanoemulsion with a non-synthetic stabilizer. During processing, samples were taken for particle size measurement and further characterization. There was a significant correlation and agreement between particle size and sensor signal as well as improved process reproducibility through sensor-guided manufacture. Given the cost associated with nanoemulsion development and scale-up manufacture, our sensor arrays could be an invaluable tool for efficient and cost-effective drug development. Sensor-guided manufacturing was used to optimize oxygen-carrying nanoemulsions. These were tested, in vitro, for their ability to improve the viability of encapsulated endocrine clusters (mouse insulinoma, Min6) and to eliminate hypoxia due to oxygen mass transfer limitations. The nanomulsions significantly improved encapsulated cluster viability and reduced hypoxia within the microcapsule environment. STATEMENT OF SIGNIFICANCE: Nanoemulsions are rapidly becoming vehicles for the controlled release delivery of both hydrophilic and hydrophobic drugs given their large surface area for exchange. As work shifts from bench to large scale manufacturing, there is a critical need for technologies that can monitor and accumulate data during processing, particularly regarding the endpoint criteria of particle size and stability. To date, no such technology has been implemented in nanoemulsion manufacture. In this paper we develop and implement an optical sensor array for in-line nanoemulsion process monitoring and then use the array to optimize the development and manufacture of novel reproducible oxygen carrying nanoemulsions lacking synthetic surfactants.
Collapse
|
8
|
Araki D, Fu JF, Huntsman H, Cordes S, Seifuddin F, Alvarado LJ, Cheruku PS, Cash A, Traba J, Li Y, Pirooznia M, Smith RH, Larochelle A. NOTCH-mediated ex vivo expansion of human hematopoietic stem and progenitor cells by culture under hypoxia. Stem Cell Reports 2021; 16:2336-2350. [PMID: 34450041 PMCID: PMC8452537 DOI: 10.1016/j.stemcr.2021.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 07/30/2021] [Accepted: 08/01/2021] [Indexed: 02/07/2023] Open
Abstract
Activation of NOTCH signaling in human hematopoietic stem/progenitor cells (HSPCs) by treatment with an engineered Delta-like ligand (DELTA1ext-IgG [DXI]) has enabled ex vivo expansion of short-term HSPCs, but the effect on long-term repopulating hematopoietic stem cells (LTR-HSCs) remains uncertain. Here, we demonstrate that ex vivo culture of human adult HSPCs with DXI under low oxygen tension limits ER stress in LTR-HSCs and lineage-committed progenitors compared with normoxic cultures. A distinct HSC gene signature was upregulated in cells cultured with DXI in hypoxia and, after 21 days of culture, the frequency of LTR-HSCs increased 4.9-fold relative to uncultured cells and 4.2-fold compared with the normoxia + DXI group. NOTCH and hypoxia pathways intersected to maintain undifferentiated phenotypes in cultured HSPCs. Our work underscores the importance of mitigating ER stress perturbations to preserve functional LTR-HSCs in extended cultures and offers a clinically feasible platform for the expansion of human HSPCs.
Collapse
Affiliation(s)
- Daisuke Araki
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Jian Fei Fu
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Heather Huntsman
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Stefan Cordes
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Fayaz Seifuddin
- Bioinformatics and Computational Biology Laboratory, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Luigi J Alvarado
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Patali S Cheruku
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Ayla Cash
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Javier Traba
- Cardiovascular Branch, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Yuesheng Li
- DNA Sequencing and Genomics Core Facility, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Mehdi Pirooznia
- Bioinformatics and Computational Biology Laboratory, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Richard H Smith
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Andre Larochelle
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), 9000 Rockville Pike, Bethesda, MD 20892, USA.
| |
Collapse
|
9
|
Tse HM, Gardner G, Dominguez-Bendala J, Fraker CA. The Importance of Proper Oxygenation in 3D Culture. Front Bioeng Biotechnol 2021; 9:634403. [PMID: 33859979 PMCID: PMC8042214 DOI: 10.3389/fbioe.2021.634403] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/09/2021] [Indexed: 12/15/2022] Open
Abstract
Cell culture typically employs inexpensive, disposable plasticware, and standard humidified CO2/room air incubators (5% CO2, ∼20% oxygen). These methods have historically proven adequate for the maintenance of viability, function, and proliferation of many cell types, but with broad variation in culture practices. With technological advances it is becoming increasingly clear that cell culture is not a “one size fits all” procedure. Recently, there is a shift toward comprehension of the individual physiological niches of cultured cells. As scale-up production of single cell and 3D aggregates for therapeutic applications has expanded, researchers have focused on understanding the role of many environmental metabolites/forces on cell function and viability. Oxygen, due to its role in cell processes and the requirement for adequate supply to maintain critical energy generation, is one such metabolite gaining increased focus. With the advent of improved sensing technologies and computational predictive modeling, it is becoming evident that parameters such as cell seeding density, culture media height, cellular oxygen consumption rate, and aggregate dimensions should be considered for experimental reproducibility. In this review, we will examine the role of oxygen in 3D cell culture with particular emphasis on primary islets of Langerhans and stem cell-derived insulin-producing SC-β cells, both known for their high metabolic demands. We will implement finite element modeling (FEM) to simulate historical and current culture methods in referenced manuscripts and innovations focusing on oxygen distribution. Our group and others have shown that oxygen plays a key role in proliferation, differentiation, and function of these 3D aggregates. Their culture in plastic consistently results in core regions of hypoxia/anoxia exacerbated by increased media height, aggregate dimensions, and oxygen consumption rates. Static gas permeable systems ameliorate this problem. The use of rotational culture and other dynamic culture systems also have advantages in terms of oxygen supply but come with the caveat that these endocrine aggregates are also exquisitely sensitive to mechanical perturbation. As recent work demonstrates, there is a strong rationale for the use of alternate in vitro systems to maintain physio-normal environments for cell growth and function for better phenotypic approximation of in vivo counterparts.
Collapse
Affiliation(s)
- Hubert M Tse
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Graeme Gardner
- Department of Surgery, Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Coral Gables, FL, United States
| | - Juan Dominguez-Bendala
- Department of Surgery, Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Coral Gables, FL, United States.,Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Christopher A Fraker
- Department of Surgery, Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Coral Gables, FL, United States
| |
Collapse
|
10
|
Molecular mechanisms of transcription factor mediated cell reprogramming: conversion of liver to pancreas. Biochem Soc Trans 2021; 49:579-590. [PMID: 33666218 PMCID: PMC8106502 DOI: 10.1042/bst20200219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/22/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022]
Abstract
Transdifferentiation is a type of cellular reprogramming involving the conversion of one differentiated cell type to another. This remarkable phenomenon holds enormous promise for the field of regenerative medicine. Over the last 20 years techniques used to reprogram cells to alternative identities have advanced dramatically. Cellular identity is determined by the transcriptional profile which comprises the subset of mRNAs, and therefore proteins, being expressed by a cell at a given point in time. A better understanding of the levers governing transcription factor activity benefits our ability to generate therapeutic cell types at will. One well-established example of transdifferentiation is the conversion of hepatocytes to pancreatic β-cells. This cell type conversion potentially represents a novel therapy in T1D treatment. The identification of key master regulator transcription factors (which distinguish one body part from another) during embryonic development has been central in developing transdifferentiation protocols. Pdx1 is one such example of a master regulator. Ectopic expression of vector-delivered transcription factors (particularly the triumvirate of Pdx1, Ngn3 and MafA) induces reprogramming through broad transcriptional remodelling. Increasingly, complimentary cell culture techniques, which recapitulate the developmental microenvironment, are employed to coax cells to adopt new identities by indirectly regulating transcription factor activity via intracellular signalling pathways. Both transcription factor-based reprogramming and directed differentiation approaches ultimately exploit transcription factors to influence cellular identity. Here, we explore the evolution of reprogramming and directed differentiation approaches within the context of hepatocyte to β-cell transdifferentiation focussing on how the introduction of new techniques has improved our ability to generate β-cells.
Collapse
|
11
|
Kouroupis D, Correa D. Increased Mesenchymal Stem Cell Functionalization in Three-Dimensional Manufacturing Settings for Enhanced Therapeutic Applications. Front Bioeng Biotechnol 2021; 9:621748. [PMID: 33644016 PMCID: PMC7907607 DOI: 10.3389/fbioe.2021.621748] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/07/2021] [Indexed: 12/23/2022] Open
Abstract
Mesenchymal stem/stromal cell (MSC) exist within their in vivo niches as part of heterogeneous cell populations, exhibiting variable stemness potential and supportive functionalities. Conventional extensive 2D in vitro MSC expansion, aimed at obtaining clinically relevant therapeutic cell numbers, results in detrimental effects on both cellular characteristics (e.g., phenotypic changes and senescence) and functions (e.g., differentiation capacity and immunomodulatory effects). These deleterious effects, added to the inherent inter-donor variability, negatively affect the standardization and reproducibility of MSC therapeutic potential. The resulting manufacturing challenges that drive the qualitative variability of MSC-based products is evident in various clinical trials where MSC therapeutic efficacy is moderate or, in some cases, totally insufficient. To circumvent these limitations, various in vitro/ex vivo techniques have been applied to manufacturing protocols to induce specific features, attributes, and functions in expanding cells. Exposure to inflammatory cues (cell priming) is one of them, however, with untoward effects such as transient expression of HLA-DR preventing allogeneic therapeutic schemes. MSC functionalization can be also achieved by in vitro 3D culturing techniques, in an effort to more closely recapitulate the in vivo MSC niche. The resulting spheroid structures provide spatial cell organization with increased cell–cell interactions, stable, or even enhanced phenotypic profiles, and increased trophic and immunomodulatory functionalities. In that context, MSC 3D spheroids have shown enhanced “medicinal signaling” activities and increased homing and survival capacities upon transplantation in vivo. Importantly, MSC spheroids have been applied in various preclinical animal models including wound healing, bone and osteochondral defects, and cardiovascular diseases showing safety and efficacy in vivo. Therefore, the incorporation of 3D MSC culturing approach into cell-based therapy would significantly impact the field, as more reproducible clinical outcomes may be achieved without requiring ex vivo stimulatory regimes. In the present review, we discuss the MSC functionalization in 3D settings and how this strategy can contribute to an improved MSC-based product for safer and more effective therapeutic applications.
Collapse
Affiliation(s)
- Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, Miami, FL, United States.,Diabetes Research Institute & Cell Transplantation Center, University of Miami, Miller School of Medicine, Miami, FL, United States
| | - Diego Correa
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, Miami, FL, United States.,Diabetes Research Institute & Cell Transplantation Center, University of Miami, Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
12
|
Patel SN, Ishahak M, Chaimov D, Velraj A, LaShoto D, Hagan DW, Buchwald P, Phelps EA, Agarwal A, Stabler CL. Organoid microphysiological system preserves pancreatic islet function within 3D matrix. SCIENCE ADVANCES 2021; 7:7/7/eaba5515. [PMID: 33579705 PMCID: PMC7880596 DOI: 10.1126/sciadv.aba5515] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 12/23/2020] [Indexed: 05/14/2023]
Abstract
Three-dimensional (3D) multicellular organoids recapitulate the native complexities of human tissue better than traditional cellular monolayers. As organoids are insufficiently supported using standard static culture, microphysiological systems (MPSs) provide a key enabling technology to maintain organoid physiology in vitro. Here, a polydimethylsiloxane-free MPS that enables continuous dynamic culture and serial in situ multiparametric assessments was leveraged to culture organoids, specifically human and rodent pancreatic islets, within a 3D alginate hydrogel. Computational modeling predicted reduced hypoxic stress and improved insulin secretion compared to static culture. Experimental validation via serial, high-content, and noninvasive assessments quantitatively confirmed that the MPS platform retained organoid viability and functionality for at least 10 days, in stark contrast to the acute decline observed overnight under static conditions. Our findings demonstrate the importance of a dynamic in vitro microenvironment for the preservation of primary organoid function and the utility of this MPS for in situ multiparametric assessment.
Collapse
Affiliation(s)
- S N Patel
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - M Ishahak
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL 33146, USA
| | - D Chaimov
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - A Velraj
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - D LaShoto
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - D W Hagan
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - P Buchwald
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - E A Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - A Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL 33146, USA.
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - C L Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
- University of Florida Diabetes Institute, Gainesville, FL 32610, USA
| |
Collapse
|
13
|
Response of Pluripotent Stem Cells to Environmental Stress and Its Application for Directed Differentiation. BIOLOGY 2021; 10:biology10020084. [PMID: 33498611 PMCID: PMC7912122 DOI: 10.3390/biology10020084] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 12/15/2022]
Abstract
Simple Summary Environmental changes in oxygen concentration, temperature, and mechanical stimulation lead to the activation of specific transcriptional factors and induce the expression of each downstream gene. In general, these responses are protective machinery against such environmental stresses, while these transcriptional factors also regulate cell proliferation, differentiation, and organ development in mammals. In the case of pluripotent stem cells, similar response mechanisms normally work and sometimes stimulate the differentiation cues. Up to now, differentiation protocols utilizing such environmental stresses have been reported to obtain various types of somatic cells from pluripotent stem cells. Basically, environmental stresses as hypoxia (low oxygen), hyperoxia, (high oxygen) and mechanical stress from cell culture plates are relatively safer than chemicals and gene transfers, which affect the genome irreversibly. Therefore, protocols designed with such environments in mind could be useful for the technology development of cell therapy and regenerative medicine. In this manuscript, we summarize recent findings of environmental stress-induced differentiation protocols and discuss their mechanisms. Abstract Pluripotent stem cells have unique characteristics compared to somatic cells. In this review, we summarize the response to environmental stresses (hypoxic, oxidative, thermal, and mechanical stresses) in embryonic stem cells (ESCs) and their applications in the differentiation methods directed to specific lineages. Those stresses lead to activation of each specific transcription factor followed by the induction of downstream genes, and one of them regulates lineage specification. In short, hypoxic stress promotes the differentiation of ESCs to mesodermal lineages via HIF-1α activation. Concerning mechanical stress, high stiffness tends to promote mesodermal differentiation, while low stiffness promotes ectodermal differentiation via the modulation of YAP1. Furthermore, each step in the same lineage differentiation favors each appropriate stiffness of culture plate; for example, definitive endoderm favors high stiffness, while pancreatic progenitor favors low stiffness during pancreatic differentiation of human ESCs. Overall, treatments utilizing those stresses have no genotoxic or carcinogenic effects except oxidative stress; therefore, the differentiated cells are safe and could be useful for cell replacement therapy. In particular, the effect of mechanical stress on differentiation is becoming attractive for the field of regenerative medicine. Therefore, the development of a stress-mediated differentiation protocol is an important matter for the future.
Collapse
|
14
|
Yang K, Lee M, Jones PA, Liu SS, Zhou A, Xu J, Sreekanth V, Wu JLY, Vo L, Lee EA, Pop R, Lee Y, Wagner BK, Melton DA, Choudhary A, Karp JM. A 3D culture platform enables development of zinc-binding prodrugs for targeted proliferation of β cells. SCIENCE ADVANCES 2020; 6:eabc3207. [PMID: 33208361 PMCID: PMC7673808 DOI: 10.1126/sciadv.abc3207] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 10/07/2020] [Indexed: 06/11/2023]
Abstract
Advances in treating β cell loss include islet replacement therapies or increasing cell proliferation rate in type 1 and type 2 diabetes, respectively. We propose developing multiple proliferation-inducing prodrugs that target high concentration of zinc ions in β cells. Unfortunately, typical two-dimensional (2D) cell cultures do not mimic in vivo conditions, displaying a markedly lowered zinc content, while 3D culture systems are laborious and expensive. Therefore, we developed the Disque Platform (DP)-a high-fidelity culture system where stem cell-derived β cells are reaggregated into thin, 3D discs within 2D 96-well plates. We validated the DP against standard 2D and 3D cultures and interrogated our zinc-activated prodrugs, which release their cargo upon zinc chelation-so preferentially in β cells. Through developing a reliable screening platform that bridges the advantages of 2D and 3D culture systems, we identified an effective hit that exhibits 2.4-fold increase in β cell proliferation compared to harmine.
Collapse
Affiliation(s)
- Kisuk Yang
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02142, USA
| | - Miseon Lee
- Chemical Biology and Therapeutics Science, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Peter Anthony Jones
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Sophie S Liu
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Angela Zhou
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jun Xu
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Vedagopuram Sreekanth
- Chemical Biology and Therapeutics Science, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Jamie L Y Wu
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lillian Vo
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Eunjee A Lee
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Ramona Pop
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Yuhan Lee
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Bridget K Wagner
- Chemical Biology and Therapeutics Science, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Douglas A Melton
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Amit Choudhary
- Chemical Biology and Therapeutics Science, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Divisions of Renal Medicine and Engineering, Brigham and Women's Hospital, Boston, MA 02115, USA
- Chemical Biology Program, Harvard University, Cambridge, MA 02138, USA
| | - Jeffrey M Karp
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
15
|
Qadir MMF, Álvarez-Cubela S, Weitz J, Panzer JK, Klein D, Moreno-Hernández Y, Cechin S, Tamayo A, Almaça J, Hiller H, Beery M, Kusmartseva I, Atkinson M, Speier S, Ricordi C, Pugliese A, Caicedo A, Fraker CA, Pastori RL, Domínguez-Bendala J. Long-term culture of human pancreatic slices as a model to study real-time islet regeneration. Nat Commun 2020; 11:3265. [PMID: 32601271 PMCID: PMC7324563 DOI: 10.1038/s41467-020-17040-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 06/04/2020] [Indexed: 01/02/2023] Open
Abstract
The culture of live pancreatic tissue slices is a powerful tool for the interrogation of physiology and pathology in an in vitro setting that retains near-intact cytoarchitecture. However, current culture conditions for human pancreatic slices (HPSs) have only been tested for short-term applications, which are not permissive for the long-term, longitudinal study of pancreatic endocrine regeneration. Using a culture system designed to mimic the physiological oxygenation of the pancreas, we demonstrate high viability and preserved endocrine and exocrine function in HPS for at least 10 days after sectioning. This extended lifespan allowed us to dynamically lineage trace and quantify the formation of insulin-producing cells in HPS from both non-diabetic and type 2 diabetic donors. This technology is expected to be of great impact for the conduct of real-time regeneration/developmental studies in the human pancreas.
Collapse
Affiliation(s)
- Mirza Muhammad Fahd Qadir
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Silvia Álvarez-Cubela
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Jonathan Weitz
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Julia K Panzer
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Dagmar Klein
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Yaisa Moreno-Hernández
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Universidad Francisco de Vitoria, Madrid, Spain
| | - Sirlene Cechin
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Alejandro Tamayo
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Joana Almaça
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Helmut Hiller
- nPOD Laboratory, Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Maria Beery
- nPOD Laboratory, Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Irina Kusmartseva
- nPOD Laboratory, Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Mark Atkinson
- nPOD Laboratory, Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32611, USA
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Stephan Speier
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany
- Faculty of Medicine, Institute of Physiology, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), München, Neuherberg, Germany
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Alberto Pugliese
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Microbiology & Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Alejandro Caicedo
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Christopher A Fraker
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Biomedical Engineering, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Ricardo Luis Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Juan Domínguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
16
|
Estrada EJ, Decima JL, Bortman G, Roberti J, Romero EB, Samaja G, Saavedra AR, Martínez G, Gutiérrez S. Combination treatment of autologous bone marrow stem cell transplantation and hyperbaric oxygen therapy for type 2 diabetes mellitus: A randomized controlled trial. Cell Transplant 2019; 28:1632-1640. [PMID: 31665912 PMCID: PMC6923554 DOI: 10.1177/0963689719883813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/17/2019] [Accepted: 09/30/2019] [Indexed: 01/06/2023] Open
Abstract
The objective of this study was to compare standard treatment versus the combination of intrapancreatic autologous stem cell (ASC) infusion and hyperbaric oxygen treatment (HBOT) before and after ASC in the metabolic control of patients with type 2 diabetes mellitus (T2DM). This study was a prospective, randomized controlled trial. The combined intervention consisted of 10 sessions of HBOT before the intrapancreatic infusion of ASC and 10 sessions afterwards. ASCs were infused into the main arterial supply of the pancreas to maximize the presence of the stem cells where the therapeutic effect is most desired. A total of 23 patients were included (control group = 10, intervention group = 13). Age, gender, diabetes duration, number of medications taken, body weight and height, and insulin requirements were recorded at baseline and every three months. Also, body mass index, fasting plasma glucose, C-peptide, and HbA1c, C-peptide/glucose ratio (CPGR) were measured every three months for one year. HbA1c was significantly lower in the intervention group compared with control throughout follow-up. Overall, 77% of patients in the intervention group and 30% of patients in the control group demonstrated a decrease of HbA1c at 180 days (compared with baseline) of at least 1 unit. Glucose levels were significantly lower in the intervention group at all timepoints during follow-up. C-peptide levels were significantly higher in the intervention group during follow-up and at one year: 1.9 ± 1.0 ng/mL versus 0.7 ± 0.4 ng/mL in intervention versus control groups, respectively, p = 0.0021. CPGR was higher in the intervention group at all controls during follow-up. The requirement for insulin was significantly lower in the intervention group at 90, 180, 270, and 365 days. Combined therapy of intrapancreatic ASC infusion and HBOT showed increased metabolic control and reduced insulin requirements in patients with T2DM compared with standard treatment.
Collapse
Affiliation(s)
- Esteban J. Estrada
- Hospital de Alta Complejidad Pte. Juan Domingo Perón, Formosa,
Argentina
| | - José Luis Decima
- Hospital de Alta Complejidad Pte. Juan Domingo Perón, Formosa,
Argentina
| | - Guillermo Bortman
- Hospital de Alta Complejidad Pte. Juan Domingo Perón, Formosa,
Argentina
| | - Javier Roberti
- Hospital de Alta Complejidad Pte. Juan Domingo Perón, Formosa,
Argentina
| | | | - Gustavo Samaja
- Hospital de Alta Complejidad Pte. Juan Domingo Perón, Formosa,
Argentina
| | | | - Gerardo Martínez
- Hospital de Alta Complejidad Pte. Juan Domingo Perón, Formosa,
Argentina
| | - Samuel Gutiérrez
- Hospital de Alta Complejidad Pte. Juan Domingo Perón, Formosa,
Argentina
| |
Collapse
|
17
|
Qadir MMF, Álvarez-Cubela S, Klein D, Lanzoni G, García-Santana C, Montalvo A, Pláceres-Uray F, Mazza EMC, Ricordi C, Inverardi LA, Pastori RL, Domínguez-Bendala J. P2RY1/ALK3-Expressing Cells within the Adult Human Exocrine Pancreas Are BMP-7 Expandable and Exhibit Progenitor-like Characteristics. Cell Rep 2019; 22:2408-2420. [PMID: 29490276 PMCID: PMC5905712 DOI: 10.1016/j.celrep.2018.02.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/08/2017] [Accepted: 02/01/2018] [Indexed: 12/16/2022] Open
Abstract
Treatment of human pancreatic non-endocrine tissue with Bone Morphogenetic Protein 7 (BMP-7) leads to the formation of glucose-responsive β-like cells. Here, we show that BMP-7 acts on extrainsular cells expressing PDX1 and the BMP receptor activin-like kinase 3 (ALK3/BMPR1A). In vitro lineage tracing indicates that ALK3+ cell populations are multipotent. PDX1+/ALK3+ cells are absent from islets but prominently represented in the major pancreatic ducts and pancreatic duct glands. We identified the purinergic receptor P2Y1 (P2RY1) as a surrogate surface marker for PDX1. Sorted P2RY1+/ALK3bright+ cells form BMP-7-expandable colonies characterized by NKX6.1 and PDX1 expression. Unlike the negative fraction controls, these colonies can be differentiated into multiple pancreatic lineages upon BMP-7 withdrawal. RNA-seq further corroborates the progenitor-like nature of P2RY1+/ALK3bright+ cells and their multilineage differentiation potential. Our studies confirm the existence of progenitor cells in the adult human pancreas and suggest a specific anatomical location within the ductal and glandular networks.
Collapse
Affiliation(s)
- Mirza Muhammad Fahd Qadir
- Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Department of Cell Biology and Anatomy, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Silvia Álvarez-Cubela
- Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Dagmar Klein
- Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Giacomo Lanzoni
- Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | | | - Abelardo Montalvo
- Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Fabiola Pláceres-Uray
- Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | | | - Camillo Ricordi
- Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Department of Microbiology & Immunology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Department of Biomedical Engineering, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Luca Alessandro Inverardi
- Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Department of Microbiology & Immunology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Ricardo Luis Pastori
- Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Department of Microbiology & Immunology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| | - Juan Domínguez-Bendala
- Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Department of Cell Biology and Anatomy, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| |
Collapse
|
18
|
Manzoli V, Colter DC, Dhanaraj S, Fornoni A, Ricordi C, Pileggi A, Tomei AA. Engineering human renal epithelial cells for transplantation in regenerative medicine. Med Eng Phys 2017; 48:3-13. [DOI: 10.1016/j.medengphy.2017.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/03/2017] [Accepted: 03/26/2017] [Indexed: 12/16/2022]
|
19
|
Landor SKJ, Lendahl U. The interplay between the cellular hypoxic response and Notch signaling. Exp Cell Res 2017; 356:146-151. [PMID: 28456549 DOI: 10.1016/j.yexcr.2017.04.030] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 01/16/2023]
Abstract
The ability to sense and adapt to low oxygen levels (hypoxia) is central for most organisms and cell types. At the center of this process is a molecular mechanism, the cellular hypoxic response, in which the hypoxia inducible factors (HIFs) are stabilized by hypoxia, allowing the HIF proteins to act as master transcriptional regulators to adjust the cell to a low oxygen environment. In recent years, it has become increasingly appreciated that the cellular hypoxic response does not always operate in splendid isolation, but intersects with signaling mechanisms such as Notch signaling, a key regulatory signaling mechanism operating in most cell types controlling stem cell maintenance and differentiation. In this review, which is dedicated to the memory of Lorenz Poellinger,1 we discuss how the intersection between Notch and the cellular hypoxic response was discovered and our current understanding of the molecular basis for the cross-talk. We also provide examples of where Notch and hypoxia intersect in various physiological and disease contexts.
Collapse
Affiliation(s)
- Sebastian K-J Landor
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Stockholm, Sweden; Department of Cell Biology, Åbo Akademi University, FI-20520 Turku, Finland
| | - Urban Lendahl
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Stockholm, Sweden; Department of Cell Biology, Åbo Akademi University, FI-20520 Turku, Finland.
| |
Collapse
|
20
|
Usman S, Khan I, Naeem N, Iqbal H, Ali A, Usman S, Salim A. Conditioned media trans-differentiate mature fibroblasts into pancreatic beta-like cells. Life Sci 2016; 164:52-59. [PMID: 27593573 DOI: 10.1016/j.lfs.2016.08.032] [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: 05/15/2016] [Revised: 08/22/2016] [Accepted: 08/31/2016] [Indexed: 10/21/2022]
Abstract
AIM The study was carried out to evaluate the role of preconditioning strategies on the trans-differentiation of mature fibroblasts (NIH3T3 cells) into insulin producing β-cells. METHODS The NIH3T3 cells were treated with dexamethasone (5μM) and pancreatic extract (0.05 and 0.4mg/mL) separately or in combination. The treated cells were analyzed for the morphological changes, and expression of pancreatic genes and proteins by phase contrast microscopy, RT-PCR and flow cytometry/immunocytochemistry, respectively. RESULTS Treatment of mature fibroblasts with different combinations of dexamethasone and pancreatic extract in the form of conditioned media resulted in comparable morphological changes and expression of certain pancreatic genes and proteins; however, their expression varied with each treatment. Most prominent effect was observed in case of combined treatment which resulted in significant increase (p<0.001) in gene expression levels of insulin, MafA, and Ngn3. Variable pattern was observed in insulin, MafA, Ngn3 and Sca1 expressions at the protein level. CONCLUSION It is concluded from this study that preconditioning of NIH3T3 cells with conditioned media containing different combinations of dexamethasone and pancreatic extract can induce trans-differentiation of these cells into pancreatic β-like cells. The conditioned media however, need to be optimized. The study may offer the possibility of improved regeneration of mature cell type that could serve as a future therapeutic option for diabetes.
Collapse
Affiliation(s)
- Shumaila Usman
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Irfan Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Nadia Naeem
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan; Dow University of Health Sciences, Ojha Campus, Gulzar-e-Hijri,Suparco Road, KDA Scheme-33, Karachi, Pakistan
| | - Hana'a Iqbal
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Anwar Ali
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan; Department of Physiology, University of Karachi, Karachi-75270, Pakistan
| | - Sehrish Usman
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan.
| |
Collapse
|
21
|
Prasadan K, Shiota C, Xiangwei X, Ricks D, Fusco J, Gittes G. A synopsis of factors regulating beta cell development and beta cell mass. Cell Mol Life Sci 2016; 73:3623-37. [PMID: 27105622 PMCID: PMC5002366 DOI: 10.1007/s00018-016-2231-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/24/2016] [Accepted: 04/14/2016] [Indexed: 12/29/2022]
Abstract
The insulin-secreting beta cells in the endocrine pancreas regulate blood glucose levels, and loss of functional beta cells leads to insulin deficiency, hyperglycemia (high blood glucose) and diabetes mellitus. Current treatment strategies for type-1 (autoimmune) diabetes are islet transplantation, which has significant risks and limitations, or normalization of blood glucose with insulin injections, which is clearly not ideal. The type-1 patients can lack insulin counter-regulatory mechanism; therefore, hypoglycemia is a potential risk. Hence, a cell-based therapy offers a better alternative for the treatment of diabetes. Past research was focused on attempting to generate replacement beta cells from stem cells; however, recently there has been an increasing interest in identifying mechanisms that will lead to the conversion of pre-existing differentiated endocrine cells into beta cells. The goal of this review is to provide an overview of several of the key factors that regulate new beta cell formation (neogenesis) and beta cell proliferation.
Collapse
Affiliation(s)
- Krishna Prasadan
- Rangos Research Center, Children's Hospital of University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Chiyo Shiota
- Rangos Research Center, Children's Hospital of University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Xiao Xiangwei
- Rangos Research Center, Children's Hospital of University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - David Ricks
- Rangos Research Center, Children's Hospital of University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Joseph Fusco
- Rangos Research Center, Children's Hospital of University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - George Gittes
- Rangos Research Center, Children's Hospital of University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.
| |
Collapse
|
22
|
Bukys MA, Bakos B, Afelik S, Zimmerman B, Barbaro B, Lin DL, Vaca P, Goldman T, Rotem A, Damaser M, Oberholzer J, Barkai U, Jensen J. Xeno-Transplantation of macro-encapsulated islets and Pluripotent Stem Cell-Derived Pancreatic Progenitors without Immunosuppression. ACTA ACUST UNITED AC 2016; 2. [PMID: 31660541 DOI: 10.19104/jorm.2017.109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Islet transplantation effectively treats diabetes but relies on immune suppression and is practically limited by the number of cadaveric islets available. An alternative cellular source is insulin-producing cells derived from pluripotent cell sources. Three animal cohorts were used in the current study to evaluate whether an oxygen-providing macro-encapsulation device, 'βAIR', could function in conjunction with human embryonic stem cells (hESCs) and their derivatives. The first cohort received macro-encapsulated undifferentiated hESCs, a second cohort received hESCs differentiated to a pancreatic progenitor state with limited endocrine differentiation. A reference cohort received human islets. Macro-encapsulation devices were implanted subcutaneously and monitored for up to 4 months. Undifferentiated pluripotent stem cells did not form teratoma but underwent cell death following implantation. Human C-peptide (hC- peptide) was detectable in host serum one week after implantation for both other cohorts. hC-peptide levels decreasing over time but remained detectable up to the end of the study. Key factors associated with mature endocrine cells were observed in grafts recovered from cohorts containing islets and hESC-derivatives including C-peptide, insulin, glucagon and urocortin 3. We conclude that the 'βAIR' macroencapsulation device is compatible with both human islets and pluripotent derivatives, but has a limited capability of sustaining undifferentiated pluripotent cells.
Collapse
Affiliation(s)
- Michael A Bukys
- Department of Stem Cell Biology and Regenerative Medicine, LRI, Cleveland Clinic Foundation
| | - Brandon Bakos
- Department of Stem Cell Biology and Regenerative Medicine, LRI, Cleveland Clinic Foundation
| | - Solomon Afelik
- Department of Stem Cell Biology and Regenerative Medicine, LRI, Cleveland Clinic Foundation
| | | | - Barbara Barbaro
- Division of Transplantation, Department of Surgery, University of Illinois at Chicago
| | - Dan Li Lin
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH
| | - Pilar Vaca
- Division of Transplantation, Department of Surgery, University of Illinois at Chicago
| | | | - Avi Rotem
- Beta-O2 Technologies, Rosh-HaAyin, Israel
| | - Margot Damaser
- Department of Stem Cell Biology and Regenerative Medicine, LRI, Cleveland Clinic Foundation.,Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH.,Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic
| | - Jose Oberholzer
- Division of Transplantation, Department of Surgery, University of Illinois at Chicago
| | | | - Jan Jensen
- Department of Stem Cell Biology and Regenerative Medicine, LRI, Cleveland Clinic Foundation
| |
Collapse
|
23
|
Conway B, Innes KE, Long D. Perfluoroalkyl substances and beta cell deficient diabetes. J Diabetes Complications 2016; 30:993-8. [PMID: 27311784 PMCID: PMC5556924 DOI: 10.1016/j.jdiacomp.2016.05.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 04/27/2016] [Accepted: 05/02/2016] [Indexed: 01/09/2023]
Abstract
AIMS Perfluoroalkyl substances (PFAS) are synthetic hydrocarbons shown to preserve pancreatic islet cell viability and reduce islet cell hypoxia and apoptosis. We investigated the relationship of serum PFAS with diabetes, and whether this varied by diabetes type. METHODS 6,460 individuals with and 60,439 without diabetes from the C8 Health Project, were categorized into three groups: type 1 (n=820), type 2 (n=4,291), or uncategorized diabetes (n=1,349, missing data on diabetes type or diabetes based on blood sugar at study entry). Four PFAS were investigated: perfluorohexane sulfonate (PFHxS), perfluorooctanoic acid (PFOA), perfluorooctane sulfonate (PFOS), and perfluorononaoic acid (PFNA). RESULTS PFAS levels were significantly lower in those with diabetes, and lowest in those with type 1 diabetes. In age and sex adjusted analyses, ORs (CI) for type 1, type 2, and uncategorized diabetes compared to no diabetes were 0.59 (0.54-0.64), 0.74 (0.71-0.77), 0.84 (0.78-0.90), respectively for PFHxS; 0.69 (0.65-0.74), 0.87 (0.89-0.91), 0.92 (0.88-0.97), respectively for PFOA; 0.65 (0.61-0.70), 0.86 (0.82-0.90), 0.93 (0.86-1.03), respectively for PFOS; and 0.65 (0.57-0.74), 0.94 (0.88-1.00), 0.95 (0.85-1.06), respectively for PFNA. Further adjustment for eGFR and other covariates did not eliminate these inverse associations. CONCLUSIONS PFAS levels were negatively associated with diabetes. This inverse relationship was strongest for type 1 diabetes, suggesting the relationship with serum PFAS may vary with the severity of islet cell deficiency.
Collapse
Affiliation(s)
- Baqiyyah Conway
- Department of Epidemiology, West Virginia University, P.O. Box 9127, Morgantown, WV 26505.
| | - Karen E Innes
- Department of Epidemiology, West Virginia University, P.O. Box 9127, Morgantown, WV 26505
| | - Dustin Long
- Department of Biostatistics, West Virginia University, P.O. Box 9127, Morgantown, WV 26505
| |
Collapse
|
24
|
Yang J, Zhou F, Xing R, Lin Y, Han Y, Teng C, Wang Q. Development of large-scale size-controlled adult pancreatic progenitor cell clusters by an inkjet-printing technique. ACS APPLIED MATERIALS & INTERFACES 2015; 7:11624-11630. [PMID: 25961432 DOI: 10.1021/acsami.5b02676] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The generation of transplantable β-cells from pancreatic progenitor cells (PPCs) could serve as an ideal cell-based therapy for diabetes. Because the transplant efficiency depends on the size of islet-like clusters, it becomes one of the key research topics to produce PPCs with controlled cluster sizes in a scalable manner. In this study, we used inkjet printing to pattern biogenic nanoparticles, i.e., mutant tobacco mosaic virus (TMV), with different spot sizes to support the formation of multicellular clusters by PPCs. We successfully achieved TMV particle patterns with variable features and sizes by adjusting the surface wettability and printing speed. The spot sizes of cell-adhesive TMV mutant arrays were in the range of 50-150 μm diameter. Mouse PPCs were seeded on the TMV-RGD (arginine-glycine-aspartate)-patterned polystyrene (PS) substrate, which consists of areas that either favor (TMV-RGD) or prohibit (bare PS) cell adhesion. The PPCs stably attached, proliferated on top of the TMV-RGD support, thus resulting in the formation of uniform and confluent PPC clusters. Furthermore, the aggregated PPCs also maintained their multipotency and were positive for E-cadherin, indicating that the formation of cell-cell junctions is critical for enhanced cell-cell contact.
Collapse
Affiliation(s)
- Jia Yang
- †State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Fang Zhou
- †State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Rubo Xing
- †State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Yuan Lin
- †State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Yanchun Han
- †State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Chunbo Teng
- §College of Life Science, Northeast Forestry University, Harbin 150040, P. R. China
| | - Qian Wang
- ∥Department of Chemistry and Biochemistry, University of South Carolina, 631 Sumter Street, Columbia, South Carolina 29208, United States
| |
Collapse
|
25
|
Gerovac BJ, Valencia M, Baumlin N, Salathe M, Conner GE, Fregien NL. Submersion and hypoxia inhibit ciliated cell differentiation in a notch-dependent manner. Am J Respir Cell Mol Biol 2014; 51:516-25. [PMID: 24754775 DOI: 10.1165/rcmb.2013-0237oc] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The epithelium that lines the conducting airways is composed of several distinct cell types that differentiate from common progenitor cells. The signals that control fate selection and differentiation of ciliated cells, a major component of the epithelium, are not completely understood. Ciliated cell differentiation can be accomplished in vitro when primary normal human bronchial epithelial (NHBE) cells are cultured at an air-liquid interface, but is inhibited when NHBE cells are cultured under submerged conditions. The mechanism by which submersion prevents ciliogenesis is not understood, but may provide clues to in vivo regulation of ciliated cell differentiation. We hypothesized that submersion creates a hypoxic environment that prevents ciliated cell differentiation by blocking the gene expression program required for ciliogenesis. This was confirmed by showing that expression of multicilin and Forkhead box J1, key factors needed for ciliated cell differentiation, was inhibited when NHBE cells were cultured in submerged and hypoxic conditions. Multicilin and Forkhead box J1 expression and ciliated cell differentiation were restored in submerged and hypoxic cells upon treatment with the γ-secretase inhibitor, N-[(3,5-difluorophenyl)acetyl]-L-alanyl-2-phenyl]glycine-1,1-dimethylethyl ester (DAPT), which suggested that Notch signaling was involved. Overexpression of Notch intracellular domain inhibited differentiation in the presence of DAPT, confirming the role of Notch signaling. These results indicate that submersion and hypoxia prevent ciliated cell differentiation by maintaining Notch signaling, which represses genes necessary for ciliogenesis. These data provide new insights into the molecular mechanisms that control human bronchial differentiation.
Collapse
|
26
|
Rymer C, Paredes J, Halt K, Schaefer C, Wiersch J, Zhang G, Potoka D, Vainio S, Gittes GK, Bates CM, Sims-Lucas S. Renal blood flow and oxygenation drive nephron progenitor differentiation. Am J Physiol Renal Physiol 2014; 307:F337-45. [PMID: 24920757 PMCID: PMC4121567 DOI: 10.1152/ajprenal.00208.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 06/04/2014] [Indexed: 12/30/2022] Open
Abstract
During kidney development, the vasculature develops via both angiogenesis (branching from major vessels) and vasculogenesis (de novo vessel formation). The formation and perfusion of renal blood vessels are vastly understudied. In the present study, we investigated the regulatory role of renal blood flow and O2 concentration on nephron progenitor differentiation during ontogeny. To elucidate the presence of blood flow, ultrasound-guided intracardiac microinjection was performed, and FITC-tagged tomato lectin was perfused through the embryo. Kidneys were costained for the vasculature, ureteric epithelium, nephron progenitors, and nephron structures. We also analyzed nephron differentiation in normoxia compared with hypoxia. At embryonic day 13.5 (E13.5), the major vascular branches were perfused; however, smaller-caliber peripheral vessels remained unperfused. By E15.5, peripheral vessels started to be perfused as well as glomeruli. While the interior kidney vessels were perfused, the peripheral vessels (nephrogenic zone) remained unperfused. Directly adjacent and internal to the nephrogenic zone, we found differentiated nephron structures surrounded and infiltrated by perfused vessels. Furthermore, we determined that at low O2 concentration, little nephron progenitor differentiation was observed; at higher O2 concentrations, more differentiation of the nephron progenitors was induced. The formation of the developing renal vessels occurs before the onset of blood flow. Furthermore, renal blood flow and oxygenation are critical for nephron progenitor differentiation.
Collapse
Affiliation(s)
- Christopher Rymer
- Rangos Research Center, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jose Paredes
- Rangos Research Center, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, and Rangos Research Center, Pittsburgh, Pennsylvania; and
| | - Kimmo Halt
- The Centre of Excellence in Cell-Extracellular Matrix Research, Oulu, Finland
| | - Caitlin Schaefer
- Rangos Research Center, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - John Wiersch
- Rangos Research Center, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, and Rangos Research Center, Pittsburgh, Pennsylvania; and
| | - Guangfeng Zhang
- Rangos Research Center, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, and Rangos Research Center, Pittsburgh, Pennsylvania; and
| | - Douglas Potoka
- Rangos Research Center, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, and Rangos Research Center, Pittsburgh, Pennsylvania; and
| | - Seppo Vainio
- The Centre of Excellence in Cell-Extracellular Matrix Research, Oulu, Finland
| | - George K Gittes
- Rangos Research Center, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, and Rangos Research Center, Pittsburgh, Pennsylvania; and
| | - Carlton M Bates
- Rangos Research Center, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sunder Sims-Lucas
- Rangos Research Center, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania;
| |
Collapse
|
27
|
White JC, Godsey ME, Bhatia SR. Perfluorocarbons enhance oxygen transport in alginate-based hydrogels. POLYM ADVAN TECHNOL 2014. [DOI: 10.1002/pat.3296] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Joseph C. White
- Department of Chemical Engineering; University of Massachusetts Amherst; Amherst MA 01003 USA
| | - Megan E. Godsey
- Department of Chemical Engineering; University of Massachusetts Amherst; Amherst MA 01003 USA
| | - Surita R. Bhatia
- Department of Chemical Engineering; University of Massachusetts Amherst; Amherst MA 01003 USA
- Department of Chemistry; Stony Brook University; Stony Brook NY 11794 USA
- Center for Functional Nanomaterials; Brookhaven National Laboratory; Upton NY 11793 USA
| |
Collapse
|
28
|
Alismail H, Jin S. Microenvironmental stimuli for proliferation of functional islet β-cells. Cell Biosci 2014; 4:12. [PMID: 24594290 PMCID: PMC3974598 DOI: 10.1186/2045-3701-4-12] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 01/29/2014] [Indexed: 12/31/2022] Open
Abstract
Diabetes is characterized by high blood glucose level due to either autoimmune destruction of islet β-cells or insufficient insulin secretion or glucose non-responsive production of insulin by β-cells. It is highly desired to replace biological functional β-cells for the treatment of diabetes. Unfortunately, β-cells proliferate with an extremely low rate. This cellular property hinders cell-based therapy for clinical application. Many attempts have been made to develop techniques that allow production of large quantities of clinically relevant islet β-cells in vitro. A line of studies evidently demonstrate that β-cells can proliferate under certain circumstances, giving the hopes for generating and expanding these cells in vitro and transplanting them to the recipient. In this review, we discuss the requirements of microenvironmental stimuli that stimulate β-cell proliferation in cell cultures. We highlight advanced approaches for augmentation of β-cell expansion that have recently emerged in this field. Furthermore, knowing the signaling pathways and molecular mechanisms would enable manipulating cell proliferation and optimizing its insulin secretory function. Thus, signaling pathways involved in the enhancement of cell proliferation are discussed as well.
Collapse
Affiliation(s)
| | - Sha Jin
- Department of Bioengineering, Thomas J, Watson School of Engineering and Applied Sciences, State University of New York in Binghamton, Binghamton, NY 13902, USA.
| |
Collapse
|
29
|
Hakim F, Kaitsuka T, Raeed JM, Wei FY, Shiraki N, Akagi T, Yokota T, Kume S, Tomizawa K. High oxygen condition facilitates the differentiation of mouse and human pluripotent stem cells into pancreatic progenitors and insulin-producing cells. J Biol Chem 2014; 289:9623-38. [PMID: 24554704 DOI: 10.1074/jbc.m113.524363] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Pluripotent stem cells have potential applications in regenerative medicine for diabetes. Differentiation of stem cells into insulin-producing cells has been achieved using various protocols. However, both the efficiency of the method and potency of differentiated cells are insufficient. Oxygen tension, the partial pressure of oxygen, has been shown to regulate the embryonic development of several organs, including pancreatic β-cells. In this study, we tried to establish an effective method for the differentiation of induced pluripotent stem cells (iPSCs) into insulin-producing cells by culturing under high oxygen (O2) conditions. Treatment with a high O2 condition in the early stage of differentiation increased insulin-positive cells at the terminus of differentiation. We found that a high O2 condition repressed Notch-dependent gene Hes1 expression and increased Ngn3 expression at the stage of pancreatic progenitors. This effect was caused by inhibition of hypoxia-inducible factor-1α protein level. Moreover, a high O2 condition activated Wnt signaling. Optimal stage-specific treatment with a high O2 condition resulted in a significant increase in insulin production in both mouse embryonic stem cells and human iPSCs and yielded populations containing up to 10% C-peptide-positive cells in human iPSCs. These results suggest that culturing in a high O2 condition at a specific stage is useful for the efficient generation of insulin-producing cells.
Collapse
Affiliation(s)
- Farzana Hakim
- From the Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Cechin S, Alvarez-Cubela S, Giraldo JA, Molano RD, Villate S, Ricordi C, Pileggi A, Inverardi L, Fraker CA, Domínguez-Bendala J. Influence of in vitro and in vivo oxygen modulation on β cell differentiation from human embryonic stem cells. Stem Cells Transl Med 2013; 3:277-89. [PMID: 24375542 DOI: 10.5966/sctm.2013-0160] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The possibility of using human embryonic stem (hES) cell-derived β cells as an alternative to cadaveric islets for the treatment of type 1 diabetes is now widely acknowledged. However, current differentiation methods consistently fail to generate meaningful numbers of mature, functional β cells. In order to address this issue, we set out to explore the role of oxygen modulation in the maturation of pancreatic progenitor (PP) cells differentiated from hES cells. We have previously determined that oxygenation is a powerful driver of murine PP differentiation along the endocrine lineage of the pancreas. We hypothesized that targeting physiological oxygen partial pressure (pO2) levels seen in mature islets would help the differentiation of PP cells along the β-cell lineage. This hypothesis was tested both in vivo (by exposing PP-transplanted immunodeficient mice to a daily hyperbaric oxygen regimen) and in vitro (by allowing PP cells to mature in a perfluorocarbon-based culture device designed to carefully adjust pO2 to a desired range). Our results show that oxygen modulation does indeed contribute to enhanced maturation of PP cells, as evidenced by improved engraftment, segregation of α and β cells, body weight maintenance, and rate of diabetes reversal in vivo, and by elevated expression of pancreatic endocrine makers, β-cell differentiation yield, and insulin production in vitro. Our studies confirm the importance of oxygen modulation as a key variable to consider in the design of β-cell differentiation protocols and open the door to future strategies for the transplantation of fully mature β cells.
Collapse
Affiliation(s)
- Sirlene Cechin
- Diabetes Research Institute, Department of Surgery, Department of Microbiology and Immunology, Department of Biomedical Engineering, Department of Medicine, and Department of Cell Biology and Anatomy, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
White JC, Stoppel WL, Roberts SC, Bhatia SR. Addition of perfluorocarbons to alginate hydrogels significantly impacts molecular transport and fracture stress. J Biomed Mater Res A 2013; 101:438-46. [PMID: 22865503 PMCID: PMC5084458 DOI: 10.1002/jbm.a.34344] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 05/04/2012] [Accepted: 05/14/2012] [Indexed: 11/11/2022]
Abstract
Perfluorocarbons (PFCs) are used in biomaterial formulations to increase oxygen (O(2) ) tension and create a homogeneous O(2) environment in three-dimensional tissue constructs. It is unclear how PFCs affect mechanical and transport properties of the scaffold, which are critical for robustness, intracellular signaling, protein transport, and overall device efficacy. In this study, we investigate composite alginate hydrogels containing a perfluorooctyl bromide (PFOB) emulsion stabilized with Pluronic(®) F68 (F68). We demonstrate that PFC addition significantly affects biomaterial properties and performance. Solution and hydrogel mechanical properties and transport of representative hydrophilic (riboflavin), hydrophobic (methyl and ethyl paraben), and protein (bovine serum albumin, BSA) solutes were compared in alginate/F68 composite hydrogels with or without PFOB. Our results indicate that mechanical properties of the alginate/F68/PFOB hydrogels are not significantly affected under small strains, but a significant decrease fracture stress is observed. The effective diffusivity D(eff) of hydrophobic small molecules decreases with PFOB emulsion addition, yet the D(eff) of hydrophilic small molecules remained unaffected. For BSA, the D(eff) increased and the loading capacity decreased with PFOB emulsion addition. Thus, a trade-off between the desired increased O(2) supply provided by PFCs and the mechanical weakening and change in transport of cellular signals must be carefully considered in the design of biomaterials containing PFCs.
Collapse
Affiliation(s)
- Joseph C. White
- Department of Chemical Engineering, University of Massachusetts Amherst, 159 Goessmann Lab, 686 North Pleasant St. Amherst, MA 01003-9303, USA
| | - Whitney L. Stoppel
- Department of Chemical Engineering, University of Massachusetts Amherst, 159 Goessmann Lab, 686 North Pleasant St. Amherst, MA 01003-9303, USA
| | - Susan C. Roberts
- Department of Chemical Engineering, University of Massachusetts Amherst, 159 Goessmann Lab, 686 North Pleasant St. Amherst, MA 01003-9303, USA
| | - Surita R. Bhatia
- Department of Chemical Engineering, University of Massachusetts Amherst, 159 Goessmann Lab, 686 North Pleasant St. Amherst, MA 01003-9303, USA
| |
Collapse
|
32
|
Benjamin S, Sheyn D, Ben-David S, Oh A, Kallai I, Li N, Gazit D, Gazit Z. Oxygenated environment enhances both stem cell survival and osteogenic differentiation. Tissue Eng Part A 2013; 19:748-58. [PMID: 23215901 DOI: 10.1089/ten.tea.2012.0298] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Osteogenesis of mesenchymal stem cells (MSCs) is highly dependent on oxygen supply. We have shown that perfluorotributylamine (PFTBA), a synthetic oxygen carrier, enhances MSC-based bone formation in vivo. Exploring this phenomenon's mechanism, we hypothesize that a transient increase in oxygen levels using PFTBA will affect MSC survival, proliferation, and differentiation, thus increasing bone formation. To test this hypothesis, MSCs overexpressing bone morphogenetic protein 2 were encapsulated in alginate beads that had been supplemented with an emulsion of PFTBA or phosphate-buffered saline. Oxygen measurements showed that supplementation of PFTBA significantly increased the available oxygen level during a 96-h period. PFTBA-containing beads displayed an elevation in cell viability, which was preserved throughout 2 weeks, and a significantly lower ratio of dead cells throughout the experiment. Furthermore, the cells from the control group expressed significantly more hypoxia-related genes such as VEGF, DDIT3, and PKG1. Additionally, PFTBA supplementation led to an increase in the osteogenic differentiation and to a decrease in chondrogenic differentiation of MSCs. In conclusion, PFTBA increases the oxygen availability in the vicinity of the MSCs, which suffer oxygen exhaustion shortly after encapsulation in alginate beads. Consequently, cell survival is increased, and hypoxia-related genes are downregulated. In addition, PFTBA promotes osteogenic differentiation over chondrogeneic differentiation, and thereby can accelerate MSC-based bone regeneration.
Collapse
Affiliation(s)
- Shimon Benjamin
- Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Gallego-Perez D, Higuita-Castro N, Reen RK, Palacio-Ochoa M, Sharma S, Lee LJ, Lannutti JJ, Hansford DJ, Gooch KJ. Micro/nanoscale technologies for the development of hormone-expressing islet-like cell clusters. Biomed Microdevices 2012; 14:779-89. [PMID: 22573223 DOI: 10.1007/s10544-012-9657-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Insulin-expressing islet-like cell clusters derived from precursor cells have significant potential in the treatment of type-I diabetes. Given that cluster size and uniformity are known to influence islet cell behavior, the ability to effectively control these parameters could find applications in the development of anti-diabetic therapies. In this work, we combined micro and nanofabrication techniques to build a biodegradable platform capable of supporting the formation of islet-like structures from pancreatic precursors. Soft lithography and electrospinning were used to create arrays of microwells (150-500 μm diameter) structurally interfaced with a porous sheet of micro/nanoscale polyblend fibers (~0.5-10 μm in cross-sectional size), upon which human pancreatic ductal epithelial cells anchored and assembled into insulin-expressing 3D clusters. The microwells effectively regulated the spatial distribution of the cells on the platform, as well as cluster size, shape and homogeneity. Average cluster cross-sectional area (~14000-17500 μm(2)) varied in proportion to the microwell dimensions, and mean circularity values remained above 0.7 for all microwell sizes. In comparison, clustering on control surfaces (fibers without microwells or tissue culture plastic) resulted in irregularly shaped/sized cell aggregates. Immunoreactivity for insulin, C-peptide and glucagon was detected on both the platform and control surfaces; however, intracellular levels of C-peptide/cell were ~60 % higher on the platform.
Collapse
Affiliation(s)
- Daniel Gallego-Perez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Fraker CA, Cechin S, Álvarez-Cubela S, Echeverri F, Bernal A, Poo R, Ricordi C, Inverardi L, Domínguez-Bendala J. A physiological pattern of oxygenation using perfluorocarbon-based culture devices maximizes pancreatic islet viability and enhances β-cell function. Cell Transplant 2012; 22:1723-33. [PMID: 23068091 DOI: 10.3727/096368912x657873] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Conventional culture vessels are not designed for physiological oxygen (O2) delivery. Both hyperoxia and hypoxia-commonly observed when culturing cells in regular plasticware-have been linked to reduced cellular function and death. Pancreatic islets, used for the clinical treatment of diabetes, are especially sensitive to sub- and supraphysiological O2 concentrations. A result of current culture standards is that a high percentage of islet preparations are never transplanted because of cell death and loss of function in the 24-48 h postisolation. Here, we describe a new culture system designed to provide quasiphysiological oxygenation to islets in culture. The use of dishes where islets rest atop a perfluorocarbon (PFC)-based membrane, coupled with a careful adjustment of environmental O2 concentration to target the islet physiological pO2 range, resulted in dramatic gains in viability and function. These observations underline the importance of approximating culture conditions as closely as possible to those of the native microenvironment, and fill a widely acknowledged gap in our ability to preserve islet functionality in vitro. As stem cell-derived insulin-producing cells are likely to suffer from the same limitations as those observed in real islets, our findings are especially timely in the context of current efforts to define renewable sources for transplantation.
Collapse
Affiliation(s)
- Chris A Fraker
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Soggia A, Flosseau K, Ravassard P, Szinnai G, Scharfmann R, Guillemain G. Activation of the transcription factor carbohydrate-responsive element-binding protein by glucose leads to increased pancreatic beta cell differentiation in rats. Diabetologia 2012; 55:2713-2722. [PMID: 22760788 PMCID: PMC3433661 DOI: 10.1007/s00125-012-2623-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 05/17/2012] [Indexed: 01/05/2023]
Abstract
AIMS/HYPOTHESIS Pancreatic cell development is a tightly controlled process. Although information is available regarding the mesodermal signals that control pancreatic development, little is known about the role of environmental factors such as nutrients, including glucose, on pancreatic development. We previously showed that glucose and its metabolism through the hexosamine biosynthesis pathway (HBP) promote pancreatic endocrine cell differentiation. Here, we analysed the role of the transcription factor carbohydrate-responsive element-binding protein (ChREBP) in this process. This transcription factor is activated by glucose, and has been recently described as a target of the HBP. METHODS We used an in vitro bioassay in which pancreatic endocrine and exocrine cells develop from rat embryonic pancreas in a way that mimics in vivo pancreatic development. Using this model, gain-of-function and loss-of-function experiments were undertaken. RESULTS ChREBP was produced in the endocrine lineage during pancreatic development, its abundance increasing with differentiation. When rat embryonic pancreases were cultured in the presence of glucose or xylitol, the production of ChREBP targets was induced. Concomitantly, beta cell differentiation was enhanced. On the other hand, when embryonic pancreases were cultured with inhibitors decreasing ChREBP activity or an adenovirus producing a dominant-negative ChREBP, beta cell differentiation was reduced, indicating that ChREBP activity was necessary for proper beta cell differentiation. Interestingly, adenovirus producing a dominant-negative ChREBP also reduced the positive effect of N-acetylglucosamine, a substrate of the HBP acting on beta cell differentiation. CONCLUSIONS/INTERPRETATION Our work supports the idea that glucose, through the transcription factor ChREBP, controls beta cell differentiation from pancreatic progenitors.
Collapse
Affiliation(s)
- A Soggia
- INSERM U845, Research Center Growth and Signalling, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Hôpital Necker, Paris, France
| | - K Flosseau
- INSERM U845, Research Center Growth and Signalling, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Hôpital Necker, Paris, France
| | - P Ravassard
- CNRS - UMR 7225, CNRS - UMR 7225 Hôpital Pitié Salpêtrière, Paris, France
| | - G Szinnai
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - R Scharfmann
- INSERM U845, Research Center Growth and Signalling, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Hôpital Necker, Paris, France
| | - G Guillemain
- INSERM U845, Research Center Growth and Signalling, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Hôpital Necker, Paris, France.
| |
Collapse
|
36
|
Cai Q, Brissova M, Reinert RB, Pan FC, Brahmachary P, Jeansson M, Shostak A, Radhika A, Poffenberger G, Quaggin SE, Jerome WG, Dumont DJ, Powers AC. Enhanced expression of VEGF-A in β cells increases endothelial cell number but impairs islet morphogenesis and β cell proliferation. Dev Biol 2012; 367:40-54. [PMID: 22546694 PMCID: PMC3391601 DOI: 10.1016/j.ydbio.2012.04.022] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 04/12/2012] [Accepted: 04/16/2012] [Indexed: 12/13/2022]
Abstract
There is a reciprocal interaction between pancreatic islet cells and vascular endothelial cells (EC) in which EC-derived signals promote islet cell differentiation and islet development while islet cell-derived angiogenic factors promote EC recruitment and extensive islet vascularization. To examine the role of angiogenic factors in the coordinated development of islets and their associated vessels, we used a "tet-on" inducible system (mice expressing rat insulin promoter-reverse tetracycline activator transgene and a tet-operon-angiogenic factor transgene) to increase the β cell production of vascular endothelial growth factor-A (VEGF-A), angiopoietin-1 (Ang1), or angiopoietin-2 (Ang2) during islet cell differentiation and islet development. In VEGF-A overexpressing embryos, ECs began to accumulate around epithelial tubes residing in the central region of the developing pancreas (associated with endocrine cells) as early as embryonic day 12.5 (E12.5) and increased dramatically by E16.5. While α and β cells formed islet cell clusters in control embryos at E16.5, the increased EC population perturbed endocrine cell differentiation and islet cell clustering in VEGF-A overexpressing embryos. With continued overexpression of VEGF-A, α and β cells became scattered, remained adjacent to ductal structures, and never coalesced into islets, resulting in a reduction in β cell proliferation and β cell mass at postnatal day 1. A similar impact on islet morphology was observed when VEGF-A was overexpressed in β cells during the postnatal period. In contrast, increased expression of Ang1 or Ang2 in β cells in developing or adult islets did not alter islet differentiation, development, or morphology, but altered islet EC ultrastructure. These data indicate that (1) increased EC number does not promote, but actually impairs β cell proliferation and islet formation; (2) the level of VEGF-A production by islet endocrine cells is critical for islet vascularization during development and postnatally; (3) angiopoietin-Tie2 signaling in endothelial cells does not have a crucial role in the development or maintenance of islet vascularization.
Collapse
Affiliation(s)
- Qing Cai
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Marcela Brissova
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Rachel B. Reinert
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Fong Cheng Pan
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Priyanka Brahmachary
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Marie Jeansson
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Canada
| | - Alena Shostak
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Aramandla Radhika
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Greg Poffenberger
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Susan E. Quaggin
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Canada
| | - W. Gray Jerome
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Daniel J. Dumont
- Division of Molecular and Cellular Biology Research, Sunnybrook Research Institute, Toronto, Ontario, Canada, M4N 3M5
| | - Alvin C. Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
| |
Collapse
|
37
|
Faleo G, Fotino C, Bocca N, Molano RD, Zahr-Akrawi E, Molina J, Villate S, Umland O, Skyler JS, Bayer AL, Ricordi C, Pileggi A. Prevention of autoimmune diabetes and induction of β-cell proliferation in NOD mice by hyperbaric oxygen therapy. Diabetes 2012; 61:1769-78. [PMID: 22566533 PMCID: PMC3379675 DOI: 10.2337/db11-0516] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
We evaluated the effects of hyperbaric oxygen therapy (HOT) on autoimmune diabetes development in nonobese diabetic (NOD) mice. Animals received no treatment or daily 60-min HOT 100% oxygen (HOT-100%) at 2.0 atmospheres absolute and were monitored for diabetes onset, insulitis, infiltrating cells, immune cell function, and β-cell apoptosis and proliferation. Cyclophosphamide-induced diabetes onset was reduced from 85.3% in controls to 48% after HOT-100% (P < 0.005) and paralleled by lower insulitis. Spontaneous diabetes incidence reduced from 85% in controls to 65% in HOT-100% (P = 0.01). Prediabetic mice receiving HOT-100% showed lower insulitis scores, reduced T-cell proliferation upon stimulation in vitro (P < 0.03), increased CD62L expression in T cells (P < 0.04), reduced costimulation markers (CD40, DC80, and CD86), and reduced major histocompatibility complex class II expression in dendritic cells (DCs) (P < 0.025), compared with controls. After autoimmunity was established, HOT was less effective. HOT-100% yielded reduced apoptosis (transferase-mediated dUTP nick-end labeling-positive insulin-positive cells; P < 0.01) and increased proliferation (bromodeoxyuridine incorporation; P < 0.001) of insulin-positive cells compared with controls. HOT reduces autoimmune diabetes incidence in NOD mice via increased resting T cells and reduced activation of DCs with preservation of β-cell mass resulting from decreased apoptosis and increased proliferation. The safety profile and noninvasiveness makes HOT an appealing adjuvant therapy for diabetes prevention and intervention trials.
Collapse
Affiliation(s)
- Gaetano Faleo
- Diabetes Research Institute, Cell Transplant Center, University of Miami, Miami, Florida
| | - Carmen Fotino
- Diabetes Research Institute, Cell Transplant Center, University of Miami, Miami, Florida
| | - Nicola Bocca
- Diabetes Research Institute, Cell Transplant Center, University of Miami, Miami, Florida
| | - R. Damaris Molano
- Diabetes Research Institute, Cell Transplant Center, University of Miami, Miami, Florida
| | - Elsie Zahr-Akrawi
- Diabetes Research Institute, Cell Transplant Center, University of Miami, Miami, Florida
| | - Judith Molina
- Diabetes Research Institute, Cell Transplant Center, University of Miami, Miami, Florida
| | - Susana Villate
- Diabetes Research Institute, Cell Transplant Center, University of Miami, Miami, Florida
| | - Oliver Umland
- Diabetes Research Institute, Cell Transplant Center, University of Miami, Miami, Florida
| | - Jay S. Skyler
- Diabetes Research Institute, Cell Transplant Center, University of Miami, Miami, Florida
- Department of Medicine, University of Miami, Miami, Florida
| | - Allison L. Bayer
- Diabetes Research Institute, Cell Transplant Center, University of Miami, Miami, Florida
- Department of Microbiology and Immunology, University of Miami, Miami, Florida
| | - Camillo Ricordi
- Diabetes Research Institute, Cell Transplant Center, University of Miami, Miami, Florida
- Department of Medicine, University of Miami, Miami, Florida
- Department of Microbiology and Immunology, University of Miami, Miami, Florida
- DeWitt Daughtry Family Department of Surgery, University of Miami, Miami, Florida
- Department of Biomedical Engineering, University of Miami, Miami, Florida
- Corresponding author: Antonello Pileggi,
| | - Antonello Pileggi
- Diabetes Research Institute, Cell Transplant Center, University of Miami, Miami, Florida
- Department of Medicine, University of Miami, Miami, Florida
- DeWitt Daughtry Family Department of Surgery, University of Miami, Miami, Florida
- Department of Biomedical Engineering, University of Miami, Miami, Florida
- Corresponding author: Antonello Pileggi,
| |
Collapse
|
38
|
Chen W, Lisowski M, Khalil G, Sweet IR, Shen AQ. Microencapsulated 3-dimensional sensor for the measurement of oxygen in single isolated pancreatic islets. PLoS One 2012; 7:e33070. [PMID: 22479359 PMCID: PMC3315556 DOI: 10.1371/journal.pone.0033070] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Accepted: 02/03/2012] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Oxygen consumption reflects multiple processes in pancreatic islets including mechanisms contributing to insulin secretion, oxidative stress and viability, providing an important readout in studies of islet function, islet viability and drug testing. Due to the scarcity, heterogeneity, and intrinsic kinetic properties of individual islets, it would be of great benefit to detect oxygen consumption by single islets. We present a novel method we have developed to image oxygen in single islets. METHODOLOGY/PRINCIPAL FINDINGS Using a microfluidics system, individual islets and a fluorescent oxygen-sensitive dye were encased within a thin alginate polymer layer. Insulin secretion by the encapsulated islets was normal. Fluorescent signal from the encased dye, detected using a standard inverted fluorescence microscope and digital camera, was stable and proportional to the amount of oxygen in the media. When integrated into a perifusion system, the sensing system detected changes in response to metabolic substrates, mitochondrial poisons, and induced-oscillations. Glucose responses averaged 30.1±7.1% of the response to a metabolic inhibitor (cyanide), increases were observed in all cases (n = 6), and the system was able to resolve changes in oxygen consumption that had a period greater than 0.5 minutes. The sensing system operated similarly from 2-48 hours following encapsulation, and viability and function of the islets were not significantly affected by the encapsulation process. CONCLUSIONS/SIGNIFICANCE An oxygen-dependent dye situated around and within a pancreatic islet encapsulated by a thin layer of alginate was sensitive to changes in oxygen consumption, and was not harmful to the function or viability of islets over the course of two days. The microcapsule-based sensing method is particularly suited to assessing the effects of compounds (dose responses and time courses) and chronic changes occurring over the course of days. The approach should be applicable to other cell types and dyes sensitive to other biologically important molecules.
Collapse
Affiliation(s)
- Wanyu Chen
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, United States of America
- School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, China
| | - Mark Lisowski
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Gamal Khalil
- Department of Aeronautics and Astronautics Department, University of Washington, Seattle, Washington, Unites States of America
| | - Ian R. Sweet
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Amy Q. Shen
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
39
|
Heinis M, Soggia A, Bechetoille C, Simon MT, Peyssonnaux C, Rustin P, Scharfmann R, Duvillié B. HIF1α and pancreatic β-cell development. FASEB J 2012; 26:2734-42. [PMID: 22426121 DOI: 10.1096/fj.11-199224] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
During early embryogenesis, the pancreas shows a paucity of blood flow, and oxygen tension, the partial pressure of oxygen (pO(2)), is low. Later, the blood flow increases as β-cell differentiation occurs. We have previously reported that pO(2) controls β-cell development in rats. Here, we checked that hypoxia inducible factor 1α (HIF1α) is essential for this control. First, we demonstrated that the effect of pO(2) on β-cell differentiation in vitro was independent of epitheliomesenchymal interactions and that neither oxidative nor energetic stress occurred. Second, the effect of pO(2) on pancreas development was shown to be conserved among species, since increasing pO(2) to 21 vs. 3% also induced β-cell differentiation in mouse (7-fold, P<0.001) and human fetal pancreas. Third, the effect of hypoxia was mediated by HIF1α, since the addition of an HIF1α inhibitor at 3% O(2) increased the number of NGN3-expressing progenitors as compared to nontreated controls (9.2-fold, P<0.001). In contrast, when we stabilized HIF1α by deleting ex vivo the gene encoding pVHL in E13.5 pancreas from Vhl floxed mice, Ngn3 expression and β-cell development decreased in such Vhl-deleted pancreas compared to controls (2.5 fold, P<0.05, and 6.6-fold, P<0.001, respectively). Taken together, these data demonstrate that HIF1α exerts a negative control over β-cell differentiation.
Collapse
Affiliation(s)
- Mylène Heinis
- Institut National de Santé et de Recherche Médicale (INSERM) U845, Research Center Growth and Signalling, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Shah SR, Esni F, Jakub A, Paredes J, Lath N, Malek M, Potoka DA, Prasadan K, Mastroberardino PG, Shiota C, Guo P, Miller KA, Hackam DJ, Burns RC, Tulachan SS, Gittes GK. Embryonic mouse blood flow and oxygen correlate with early pancreatic differentiation. Dev Biol 2010; 349:342-9. [PMID: 21050843 DOI: 10.1016/j.ydbio.2010.10.033] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 09/27/2010] [Accepted: 10/25/2010] [Indexed: 11/19/2022]
Abstract
The mammalian embryo represents a fundamental paradox in biology. Its location within the uterus, especially early during development when embryonic cardiovascular development and placental blood flow are not well-established, leads to an obligate hypoxic environment. Despite this hypoxia, the embryonic cells are able to undergo remarkable growth, morphogenesis, and differentiation. Recent evidence suggests that embryonic organ differentiation, including pancreatic β-cells, is tightly regulated by oxygen levels. Since a major determinant of oxygen tension in mammalian embryos after implantation is embryonic blood flow, here we used a novel survivable in utero intracardiac injection technique to deliver a vascular tracer to living mouse embryos. Once injected, the embryonic heart could be visualized to continue contracting normally, thereby distributing the tracer specifically only to those regions where embryonic blood was flowing. We found that the embryonic pancreas early in development shows a remarkable paucity of blood flow and that the presence of blood flow correlates with the differentiation state of the developing pancreatic epithelial cells in the region of the blood flow.
Collapse
Affiliation(s)
- Sohail R Shah
- Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, 45th Street and Penn Avenue, Pittsburgh, PA 15201, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Heinis M, Simon M, Duvillié B. New insights into endocrine pancreatic development: the role of environmental factors. Horm Res Paediatr 2010; 74:77-82. [PMID: 20551619 PMCID: PMC3202916 DOI: 10.1159/000314894] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 05/05/2010] [Indexed: 01/06/2023] Open
Abstract
The pancreas is a mixed gland that contains endocrine and exocrine components. Within the pancreatic islets, beta cells produce insulin and control the glycemia. Their deficiency leads to diabetes and several potential complications. In the last decade, numerous studies have focused on pancreas development. The objective was to characterize the cellular and molecular factors that control the differentiation of endocrine and exocrine cell types. Investigation of the role of transcription factors by using genetic approaches led to the discovery of key molecules that are expressed both in rodents and humans. Some of them are ubiquitous, and some others are specifically involved in endocrine or exocrine specification. In addition to these intrinsic factors, recent studies have focused on the role of environmental factors. In the present review, we describe the roles of nutrients and oxygen in the embryonic pancreas. Interestingly, these extrinsic parameters can interfere with beta-cell differentiation and function. Altogether, these data should help to generate beta cells in vitro and define strategies for a cell-based therapy of type 1 diabetes.
Collapse
Affiliation(s)
- M. Heinis
- INSERM U845, Research Center Growth and Signalling, Université Paris Descartes, Faculté de Médecine, Hôpital Necker, Paris, France
| | - M.T. Simon
- INSERM U845, Research Center Growth and Signalling, Université Paris Descartes, Faculté de Médecine, Hôpital Necker, Paris, France
| | - B. Duvillié
- *Dr. Bertrand Duvillié, U845 INSERM, Faculty Necker, 156, rue de Vaugirard, FR–75015 Paris (France), Tel. +33 1 40 61 55 71, Fax +33 1 43 06 04 43, E-Mail
| |
Collapse
|
42
|
Fujita H, Shimizu K, Morioka Y, Nagamori E. Enhancement of C2C12 differentiation by perfluorocarbon-mediated oxygen delivery. J Biosci Bioeng 2010; 110:359-62. [PMID: 20547328 DOI: 10.1016/j.jbiosc.2010.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 02/19/2010] [Accepted: 02/22/2010] [Indexed: 01/08/2023]
Abstract
We have studied the effect of enhanced oxygen delivery by perfluorocarbons on the differentiation of C2C12 cells. The extent of differentiation was assessed by means of phase contrast/fluorescence microscopy, active tension measurement and the glucose consumption/lactate production rates. We found that enhanced oxygen delivery is suitable for full differentiation of C2C12 cells.
Collapse
Affiliation(s)
- Hideaki Fujita
- Toyota Central Research and Development Labs., Inc., 41-1 Yokomichi, Nagakute, Aichi 480-1192, Japan
| | | | | | | |
Collapse
|
43
|
Heinis M, Simon MT, Ilc K, Mazure NM, Pouysségur J, Scharfmann R, Duvillié B. Oxygen tension regulates pancreatic beta-cell differentiation through hypoxia-inducible factor 1alpha. Diabetes 2010; 59:662-9. [PMID: 20009089 PMCID: PMC2828660 DOI: 10.2337/db09-0891] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Recent evidence indicates that low oxygen tension (pO2) or hypoxia controls the differentiation of several cell types during development. Variations of pO2 are mediated through the hypoxia-inducible factor (HIF), a crucial mediator of the adaptative response of cells to hypoxia. The aim of this study was to investigate the role of pO2 in beta-cell differentiation. RESEARCH DESIGN AND METHODS We analyzed the capacity of beta-cell differentiation in the rat embryonic pancreas using two in vitro assays. Pancreata were cultured either in collagen or on a filter at the air/liquid interface with various pO2. An inhibitor of the prolyl hydroxylases, dimethyloxaloylglycine (DMOG), was used to stabilize HIF1alpha protein in normoxia. RESULTS When cultured in collagen, embryonic pancreatic cells were hypoxic and expressed HIF1alpha and rare beta-cells differentiated. In pancreata cultured on filter (normoxia), HIF1alpha expression decreased and numerous beta-cells developed. During pancreas development, HIF1alpha levels were elevated at early stages and decreased with time. To determine the effect of pO2 on beta-cell differentiation, pancreata were cultured in collagen at increasing concentrations of O2. Such conditions repressed HIF1alpha expression, fostered development of Ngn3-positive endocrine progenitors, and induced beta-cell differentiation by O2 in a dose-dependent manner. By contrast, forced expression of HIF1alpha in normoxia using DMOG repressed Ngn3 expression and blocked beta-cell development. Finally, hypoxia requires hairy and enhancer of split (HES)1 expression to repress beta-cell differentiation. CONCLUSIONS These data demonstrate that beta-cell differentiation is controlled by pO2 through HIF1alpha. Modifying pO2 should now be tested in protocols aiming to differentiate beta-cells from embryonic stem cells.
Collapse
Affiliation(s)
- Mylène Heinis
- INSERM U845, Research Center Growth and Signalling, Université Paris Descartes, Hôpital Necker, Paris, France
| | - Marie-Thérèse Simon
- INSERM U845, Research Center Growth and Signalling, Université Paris Descartes, Hôpital Necker, Paris, France
| | - Karine Ilc
- Institute of Developmental Biology and Cancer Research, University of Nice, Nice, France
| | - Nathalie M. Mazure
- Institute of Developmental Biology and Cancer Research, University of Nice, Nice, France
| | - Jacques Pouysségur
- Institute of Developmental Biology and Cancer Research, University of Nice, Nice, France
| | - Raphael Scharfmann
- INSERM U845, Research Center Growth and Signalling, Université Paris Descartes, Hôpital Necker, Paris, France
| | - Bertrand Duvillié
- INSERM U845, Research Center Growth and Signalling, Université Paris Descartes, Hôpital Necker, Paris, France
- Corresponding author: Bertrand Duvillié,
| |
Collapse
|
44
|
The effects of low oxygen on self-renewal and differentiation of embryonic stem cells. Curr Opin Organ Transplant 2009; 14:694-700. [DOI: 10.1097/mot.0b013e3283329d53] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
45
|
The use of a synthetic oxygen carrier-enriched hydrogel to enhance mesenchymal stem cell-based bone formation in vivo. Biomaterials 2009; 30:4639-48. [PMID: 19540585 DOI: 10.1016/j.biomaterials.2009.05.027] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2009] [Accepted: 05/15/2009] [Indexed: 11/20/2022]
Abstract
A major hurdle to surmount in bone-tissue engineering is ensuring a sufficient oxygen supply to newly forming tissue to avoid cell death or delayed development of osteogenic features. We hypothesized that an oxygen-enriched hydrogel scaffold would enhance tissue-engineered bone formation in vivo. To test this, we used a well-characterized mesenchymal stem cell (MSC) line, Tet-off BMP2 MSC, whose cells were engineered to express recombinant human bone morphogenetic protein-2. Cells were suspended in hydrogel supplemented with perfluorotributylamine (PFTBA) and implanted subcutaneously in an ectopic site, a radial bone defect, or a lumbar paravertebral muscle (mouse model of spinal fusion) in C3H/HeN mice. For controls, we used cells suspended in the same gel without PFTBA. In the ectopic site, there were significant increases in bone formation (2.5-fold increase), cell survival, and osteocalcin activity in the PFTBA-supplemented groups. PFTBA supplementation significantly increased structural parameters of bone in radial bone defects and triggered a significant 1.4-fold increase in bone volume in the spinal fusion model. We conclude that synthetic oxygen carrier supplementation of tissue-engineered implants enhances ectopic bone formation and yields better bone quality and volume in bone-repair and spinal fusion models, probably due to increased cell survival.
Collapse
|
46
|
Abstract
Beyond its role as an electron acceptor in aerobic respiration, oxygen is also a key effector of many developmental events. The oxygen-sensing machinery and the very fabric of cell identity and function have been shown to be deeply intertwined. Here we take a first look at how oxygen might lie at the crossroads of at least two of the major molecular pathways that shape pancreatic development. Based on recent evidence and a thorough review of the literature, we present a theoretical model whereby evolving oxygen tensions might choreograph to a large extent the sequence of molecular events resulting in the development of the organ. In particular, we propose that lower oxygenation prior to the expansion of the vasculature may favour HIF (hypoxia inducible factor)-mediated activation of Notch and repression of Wnt/beta-catenin signalling, limiting endocrine cell differentiation. With the development of vasculature and improved oxygen delivery to the developing organ, HIF-mediated support for Notch signalling may decline while the beta-catenin-directed Wnt signalling is favoured, which would support endocrine cell differentiation and perhaps exocrine cell proliferation/differentiation.
Collapse
|
47
|
Buchwald P. FEM-based oxygen consumption and cell viability models for avascular pancreatic islets. Theor Biol Med Model 2009; 6:5. [PMID: 19371422 PMCID: PMC2678100 DOI: 10.1186/1742-4682-6-5] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Accepted: 04/16/2009] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The function and viability of cultured, transplanted, or encapsulated pancreatic islets is often limited by hypoxia because these islets have lost their vasculature during the isolation process and have to rely on gradient-driven passive diffusion, which cannot provide adequate oxygen transport. Pancreatic islets (islets of Langerhans) are particularly susceptible due to their relatively large size, large metabolic demand, and increased sensitivity to hypoxia. Here, finite element method (FEM) based multiphysics models are explored to describe oxygen transport and cell viability in avascular islets both in static and in moving culture media. METHODS Two- and three-dimensional models were built in COMSOL Multiphysics using the convection and diffusion as well as the incompressible Navier-Stokes fluid dynamics application modes. Oxygen consumption was assumed to follow Michaelis-Menten-type kinetics and to cease when local concentrations fell below a critical threshold; in a dynamic model, it was also allowed to increase with increasing glucose concentration. RESULTS Partial differential equation (PDE) based exploratory cellular-level oxygen consumption and cell viability models incorporating physiologically realistic assumptions have been implemented for fully scaled cell culture geometries with 100, 150, and 200 microm diameter islets as representative. Calculated oxygen concentrations and intra-islet regions likely to suffer from hypoxia-related necrosis obtained for traditional flask-type cultures, oxygen-permeable silicone-rubber membrane bottom cultures, and perifusion chambers with flowing media and varying incoming glucose levels are presented in detail illustrated with corresponding colour-coded figures and animations. CONCLUSION Results of the computational models are, as a first estimate, in good quantitative agreement with existing experimental evidence, and they confirm that during culture, hypoxia is often a problem for non-vascularised islet and can lead to considerable cell death (necrosis), especially in the core region of larger islets. Such models are of considerable interest to improve the function and viability of cultured, transplanted, or encapsulated islets. The present implementation allows convenient extension to true multiphysics applications that solve coupled physics phenomena such as diffusion and consumption with convection due to flowing or moving media.
Collapse
Affiliation(s)
- Peter Buchwald
- Diabetes Research Institute and the Department of Molecular and Cellular Pharmacology, University of Miami, Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
48
|
Hallén S, Clapham JC. Cell based in vitro and ex vivo models in metabolic disease drug discovery: nice to have or critical path? Expert Opin Drug Discov 2009; 4:417-28. [PMID: 23485042 DOI: 10.1517/17460440902821640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND The use of cellular models as tools in drug discovery is almost universal. However, in disease areas such as metabolic diseases, are they relevant to the process and do they add value? OBJECTIVE In this article, we explore the variety of cellular models now used in drug discovery in metabolic diseases as revealed by publication. We have tried to make some connections between drug phenotypes in these models with clinical parallels. We also ask the question as to whether such models add value in the drug discovery process. This overview is not about recombinant cell systems used in target-based screening; rather, we focus on in vitro, including ex vivo, models as physiological systems in drug discovery in obesity and diabetes. CONCLUSION In terms of building target confidence, in vitro models are often the only mechanistic link to human systems early in a projects life. Many of the current targets in metabolic diseases in the early discovery phase are not yet clinically supported, let alone validated. In this respect, therefore, in vitro models warrant a place in the critical path in early discovery. In terms of any predictive role for decision-making today, this is much more difficult and is more likely pushed to a supporting role as part of a wider package. However, there is a rapid rate of advancement in this field and future developments hold much promise.
Collapse
Affiliation(s)
- Stefan Hallén
- Departments of Bioscience, AstraZeneca R&D Mölndal, Sweden +46 31 7064339 ; +46 31 7763700 ;
| | | |
Collapse
|
49
|
Aslanidi KB, Myakisheva SN, Ivanitsky GR. Ionic regulation of proliferation of NIE-115 mouse neuroblastoma cells in vitro. DOKL BIOCHEM BIOPHYS 2009; 423:331-3. [PMID: 19230381 DOI: 10.1134/s1607672908060033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- K B Aslanidi
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, pr Nauki 3, Pushchino, Moscow oblast, 142290 Russia
| | | | | |
Collapse
|
50
|
Ichii H, Ricordi C. Current status of islet cell transplantation. ACTA ACUST UNITED AC 2008; 16:101-12. [PMID: 19110649 DOI: 10.1007/s00534-008-0021-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2008] [Accepted: 07/15/2008] [Indexed: 02/08/2023]
Abstract
Despite substantial advances in islet isolation methods and immunosuppressive protocol, pancreatic islet cell transplantation remains an experimental procedure currently limited to the most severe cases of type 1 diabetes mellitus. The objectives of this treatment are to prevent severe hypoglycemic episodes in patients with hypoglycemia unawareness and to achieve a more physiological metabolic control. Insulin independence and long term-graft function with improvement of quality of life have been obtained in several international islet transplant centers. However, experimental trials of islet transplantation clearly highlighted several obstacles that remain to be overcome before the procedure could be proposed to a much larger patient population. This review provides a brief historical perspective of islet transplantation, islet isolation techniques, the transplant procedure, immunosuppressive therapy, and outlines current challenges and future directions in clinical islet transplantation.
Collapse
Affiliation(s)
- Hirohito Ichii
- Cell Transplant Center, Diabetes Research Institute, University of Miami Leonard M Miller School of Medicine, Miami, FL 33136, USA.
| | | |
Collapse
|