1
|
Ebrahim N, Kondratyev N, Artyuhov A, Timofeev A, Gurskaya N, Andrianov A, Izrailov R, Volchkov E, Dyuzheva T, Kopantseva E, Kiseleva E, Golimbet V, Dashinimaev E. Human pancreatic islet-derived stromal cells reveal combined features of mesenchymal stromal cells and pancreatic stellate cells. Stem Cell Res Ther 2024; 15:351. [PMID: 39380125 PMCID: PMC11463112 DOI: 10.1186/s13287-024-03963-2] [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: 08/16/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) are recognized for their potential in regenerative medicine, attributed to their multipotent differentiation capabilities and immunomodulatory properties. Despite this potential, the classification and detailed characterization of MSCs, especially those derived from specific tissues like the pancreas, remains challenging leading to a proliferation of terminology in the literature. This study aims to address these challenges by providing a thorough characterization of human pancreatic islets-derived mesenchymal stromal cells (hPD-MSCs). METHODS hPD-MSCs were isolated from donor islets using enzymatic digestion, immortalized through lentiviral transduction of human telomerase reverse transcriptase (hTERT). Cells were characterized by immunostaining, flow cytometry and multilineage differentiation potential into adipogenic and osteogenic lineages. Further a transcriptomic analysis was done to compare the gene expression profiles of hPD-MSCs with other mesenchymal cells. RESULTS We show that hPD-MSCs express the classical MSC features, including morphological characteristics, surface markers expression (CD90, CD73, CD105, CD44, and CD106) and the ability to differentiate into both adipogenic and osteogenic lineages. Furthermore, transcriptomic analysis revealed distinct gene expression profiles, showing notable similarities between hPD-MSCs and pancreatic stellate cells (PSCs). The study also identified specific genes that distinguish hPD-MSCs from MSCs of other origins, including genes associated with pancreatic function (e.g., ISL1) and neural development (e.g., NPTX1, ZNF804A). A novel gene with an unknown function (ENSG00000286190) was also discovered. CONCLUSIONS This study enhances the understanding of hPD-MSCs, demonstrating their unique characteristics and potential applications in therapeutic strategies. The identification of specific gene expression profiles differentiates hPD-MSCs from other mesenchymal cells and opens new avenues for research into their role in pancreatic function and neural development.
Collapse
Affiliation(s)
- Nour Ebrahim
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia, 117997
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia, 141701
| | | | - Alexander Artyuhov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia, 117997
- Research Institute of Molecular and Cellular Medicine, RUDN University, Moscow, Russia, 117198
| | - Alexei Timofeev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia, 117997
| | - Nadya Gurskaya
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia, 117997
| | - Alexey Andrianov
- Loginov Moscow Clinical Scientific Center, Moscow, Russia, 111123
| | - Roman Izrailov
- Loginov Moscow Clinical Scientific Center, Moscow, Russia, 111123
| | - Egor Volchkov
- Research Institute of Molecular and Cellular Medicine, RUDN University, Moscow, Russia, 117198
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology (D. Rogachev, NMRCPHOI) of Ministry of Healthcare of the Russian Federation, 1, Samory Mashela St, Moscow, Russia, 117997
| | - Tatyana Dyuzheva
- Department of Hospital Surgery, Sklifosovsky Institute for Clinical Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia, 119435
| | - Elena Kopantseva
- Research Institute of Molecular and Cellular Medicine, RUDN University, Moscow, Russia, 117198
| | - Ekaterina Kiseleva
- Research Institute for Systems Biology and Medicine, Moscow, Russia, 117246
| | - Vera Golimbet
- Mental Health Research Center, Moscow, Russia, 115522
| | - Erdem Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia, 117997.
- Research Institute of Molecular and Cellular Medicine, RUDN University, Moscow, Russia, 117198.
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia, 141701.
- Institute of Medicine, Banzarov Buryat State University, Ulan-Ude, Russia, 670000.
| |
Collapse
|
2
|
Zhang Y, Yi Y, Xiao X, Hu L, Xu J, Zheng D, Koc HC, Chan UI, Meng Y, Lu L, Liu W, Xu X, Shao N, Cheung ECW, Xu RH, Chen G. Definitive Endodermal Cells Supply an in vitro Source of Mesenchymal Stem/Stromal Cells. Commun Biol 2023; 6:476. [PMID: 37127734 PMCID: PMC10151361 DOI: 10.1038/s42003-023-04810-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/05/2023] [Indexed: 05/03/2023] Open
Abstract
Mesenchymal stem/Stromal cells (MSCs) have great therapeutic potentials, and they have been isolated from various tissues and organs including definitive endoderm (DE) organs, such as the lung, liver and intestine. MSCs have been induced from human pluripotent stem cells (hPSCs) through multiple embryonic lineages, including the mesoderm, neural crest, and extraembryonic cells. However, it remains unclear whether hPSCs could give rise to MSCs in vitro through the endodermal lineage. Here, we report that hPSC-derived, SOX17+ definitive endoderm progenitors can further differentiate to cells expressing classic MSC markers, which we name definitive endoderm-derived MSCs (DE-MSCs). Single cell RNA sequencing demonstrates the stepwise emergence of DE-MSCs, while endoderm-specific gene expression can be elevated by signaling modulation. DE-MSCs display multipotency and immunomodulatory activity in vitro and possess therapeutic effects in a mouse ulcerative colitis model. This study reveals that, in addition to the other germ layers, the definitive endoderm can also contribute to MSCs and DE-MSCs could be a cell source for regenerative medicine.
Collapse
Affiliation(s)
- Yumeng Zhang
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Ye Yi
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Xia Xiao
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Lingling Hu
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Jiaqi Xu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Dejin Zheng
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Ho Cheng Koc
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Un In Chan
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Ya Meng
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Zhuhai Precision Medical Center, Zhuhai People's Hospital, Jinan University, Zhuhai, Guangdong, China
| | - Ligong Lu
- Zhuhai Precision Medical Center, Zhuhai People's Hospital, Jinan University, Zhuhai, Guangdong, China
| | - Weiwei Liu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Biological Imaging and Stem Cell Core Facility, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
| | - Xiaoling Xu
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
| | - Ningyi Shao
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
| | - Edwin Chong Wing Cheung
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
| | - Ren-He Xu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China.
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China.
| | - Guokai Chen
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China.
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China.
| |
Collapse
|
3
|
Montanari E, Szabó L, Balaphas A, Meyer J, Perriraz-Mayer N, Pimenta J, Giraud MN, Egger B, Gerber-Lemaire S, Bühler L, Gonelle-Gispert C. Multipotent mesenchymal stromal cells derived from porcine exocrine pancreas improve insulin secretion from juvenile porcine islet cell clusters. Xenotransplantation 2021; 28:e12666. [PMID: 33538027 DOI: 10.1111/xen.12666] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/30/2020] [Accepted: 11/26/2020] [Indexed: 01/03/2023]
Abstract
Neonatal and juvenile porcine islet cell clusters (ICC) present an unlimited source for islet xenotransplantation to treat type 1 diabetes patients. We isolated ICC from pancreata of 14 days old juvenile piglets and characterized their maturation by immunofluorescence and insulin secretion assays. Multipotent mesenchymal stromal cells derived from exocrine tissue of same pancreata (pMSC) were characterized for their differentiation potential and ability to sustain ICC insulin secretion in vitro and in vivo. Isolation of ICC resulted in 142 ± 50 × 103 IEQ per pancreas. Immunofluorescence staining revealed increasing presence of insulin-positive beta cells between day 9 and 21 in culture and insulin content per 500IEC of ICC increased progressively over time from 1178.4 ± 450 µg/L to 4479.7 ± 1954.2 µg/L from day 7 to 14, P < .001. Highest glucose-induced insulin secretion by ICC was obtained at day 7 of culture and reached a fold increase of 2.9 ± 0.4 compared to basal. Expansion of adherent cells from the pig exocrine tissue resulted in a homogenous CD90+ , CD34- , and CD45- fibroblast-like cell population and differentiation into adipocytes and chondrocytes demonstrated their multipotency. Insulin release from ICC was increased in the presence of pMSC and dependent on cell-cell contact (glucose-induced fold increase: ICC alone: 1.6 ± 0.2; ICC + pMSC + contact: 3.2 ± 0.5, P = .0057; ICC + pMSC no-contact: 1.9 ± 0.3; theophylline stimulation: alone: 5.4 ± 0.7; pMSC + contact: 8.4 ± 0.9, P = .013; pMSC no-contact: 5.2 ± 0.7). After transplantation of encapsulated ICC using Ca2+ -alginate (alg) microcapsules into streptozotocin-induced diabetic and immunocompetent mice, transient normalization of glycemia was obtained up to day 7 post-transplant, whereas ICC co-encapsulated with pMSC did not improve glycemia and showed increased pericapsular fibrosis. We conclude that pMSC derived from juvenile porcine exocrine pancreas improves insulin secretion of ICC by direct cell-cell contact. For transplantation purposes, the use of pMSC to support beta-cell function will depend on the development of new anti-fibrotic polymers and/or on genetically modified pigs with lower immunogenicity.
Collapse
Affiliation(s)
- Elisa Montanari
- Surgical Research Unit, CMU-1, University Hospitals of Geneva, Geneva, Switzerland
| | - Luca Szabó
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, EPFL SB ISIC SCI-SB-SG, Lausanne, Switzerland
| | - Alexandre Balaphas
- Surgical Research Unit, CMU-1, University Hospitals of Geneva, Geneva, Switzerland
| | - Jeremy Meyer
- Surgical Research Unit, CMU-1, University Hospitals of Geneva, Geneva, Switzerland
| | - Nadja Perriraz-Mayer
- Surgical Research Unit, CMU-1, University Hospitals of Geneva, Geneva, Switzerland
| | - Joel Pimenta
- Surgical Research Unit, CMU-1, University Hospitals of Geneva, Geneva, Switzerland
| | - Marie-Noelle Giraud
- Cardiology, Dpt EMC, Section of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Bernhard Egger
- Surgical Research Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Sandrine Gerber-Lemaire
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, EPFL SB ISIC SCI-SB-SG, Lausanne, Switzerland
| | - Leo Bühler
- Surgical Research Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Carmen Gonelle-Gispert
- Surgical Research Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
4
|
Garcia PE, Scales MK, Allen BL, Pasca di Magliano M. Pancreatic Fibroblast Heterogeneity: From Development to Cancer. Cells 2020; 9:E2464. [PMID: 33198201 PMCID: PMC7698149 DOI: 10.3390/cells9112464] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/10/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is characterized by an extensive fibroinflammatory microenvironment that accumulates from the onset of disease progression. Cancer-associated fibroblasts (CAFs) are a prominent cellular component of the stroma, but their role during carcinogenesis remains controversial, with both tumor-supporting and tumor-restraining functions reported in different studies. One explanation for these contradictory findings is the heterogeneous nature of the fibroblast populations, and the different roles each subset might play in carcinogenesis. Here, we review the current literature on the origin and function of pancreatic fibroblasts, from the developing organ to the healthy adult pancreas, and throughout the initiation and progression of PDA. We also discuss clinical approaches to targeting fibroblasts in PDA.
Collapse
Affiliation(s)
- Paloma E. Garcia
- Program in Molecular and Cellular Pathology, University of Michigan Medical School, University of Michigan, Ann Arbor, MI 48105, USA;
| | - Michael K. Scales
- Department of Cell and Developmental Biology, University of Michigan Medical School, University of Michigan, Ann Arbor, MI 48109, USA; (M.K.S.); (B.L.A.)
| | - Benjamin L. Allen
- Department of Cell and Developmental Biology, University of Michigan Medical School, University of Michigan, Ann Arbor, MI 48109, USA; (M.K.S.); (B.L.A.)
- Rogel Cancer Center, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marina Pasca di Magliano
- Department of Cell and Developmental Biology, University of Michigan Medical School, University of Michigan, Ann Arbor, MI 48109, USA; (M.K.S.); (B.L.A.)
- Rogel Cancer Center, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
5
|
Hong Y, Park EY, Kim D, Lee H, Jung HS, Jun HS. Glucosamine potentiates the differentiation of adipose-derived stem cells into glucose-responsive insulin-producing cells. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:561. [PMID: 32775362 PMCID: PMC7347784 DOI: 10.21037/atm.2020.03.103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background Islet transplantation might be a logical strategy to restore insulin secretion for the treatment of diabetes, however, the scarcity of donors poses an obstacle for such a treatment. As an alternative islet source, differentiation of stem cells into insulin-producing cells (IPCs) has been tried. Many protocols have been developed to improve the efficiency of differentiation of stem cells into IPCs. In this study, we investigated whether glucosamine supplementation during differentiation of human adipose-derived stem cells (hADSCs) into IPCs can improve the insulin secretory function. Methods Glucosamine was added to the original differentiation medium at different stages of differentiation of hADSCs into IPCs for 12 days and insulin secretion was analyzed. Results Addition of glucosamine alone to the growth medium of hADSCs did not affect the differentiation of hADSCs to IPCs. Supplementation of the differentiation medium with glucosamine at a later stage (protocol G3) proved to have the greatest effect on IPC differentiation. Basal and glucose-stimulated insulin secretion (GSIS) was significantly increased and the expression of insulin and C-peptide was increased in differentiated IPCs as compared with that in differentiated IPCs using the conventional protocol (protocol C). In addition, the expression of beta-cell specific transcription factors such as pancreatic and duodenal homeobox1 (PDX1) and neurogenin 3 (NGN3) was also increased. Furthermore, the expression of genes related to insulin secretion, including synaptotagmin 4 (Syt4), glucokinase (Gck) and glucose transporter 2 (Glut2), was also increased. Conclusions We conclude that glucosamine supplementation potentiates the differentiation of hADSCs into IPCs.
Collapse
Affiliation(s)
- Yeonhee Hong
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, Republic of Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Eun-Young Park
- College of Pharmacy, Mokpo National University, Muan-gun, Jeonnam, Republic of Korea
| | - Donghee Kim
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Hakmo Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hye Seung Jung
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hee-Sook Jun
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, Republic of Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea.,Gachon Medical Research Institute, Gil Hospital, Incheon, Republic of Korea
| |
Collapse
|
6
|
Cooper TT, Sherman SE, Bell GI, Ma J, Kuljanin M, Jose SE, Lajoie GA, Hess DA. Characterization of a Vimentin high /Nestin high proteome and tissue regenerative secretome generated by human pancreas-derived mesenchymal stromal cells. Stem Cells 2020; 38:666-682. [PMID: 31904137 DOI: 10.1002/stem.3143] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/14/2019] [Indexed: 12/11/2022]
Abstract
Multipotent/mesenchymal stromal cells (MSCs) exist within a variety of postnatal tissues; however, global proteomic analyses comparing tissue-specific MSC are limited. Using human bone marrow (BM)-derived MSCs as a gold standard, we used label-free mass spectrometry and functional assays to characterize the proteome, secretome, and corresponding function of human pancreas-derived MSCs (Panc-MSCs) with a classical phenotype (CD90+/CD73+/CD105+/CD45-/CD31-). Both MSC subtypes expressed mesenchymal markers vimentin, α-SMA, and STRO-1; however, expression of nestin was increased in Panc-MSCs. Accordingly, these Vimentinhigh /Nestinhigh cells were isolated from fresh human pancreatic islet and non-islet tissues. Next, we identified expression of >60 CD markers shared between Panc-MSCs and BM-MSCs, including validated expression of CD14. An additional 19 CD markers were differentially expressed, including reduced pericyte-marker CD146 expression on Panc-MSCs. Panc-MSCs also showed reduced expression of proteins involved in lipid and retinoid metabolism. Accordingly, Panc-MSCs showed restricted responses to adipogenic stimuli in vitro, although both MSC types demonstrated trilineage differentiation. In contrast, Panc-MSCs demonstrated accelerated growth kinetics and competency to pro-neurogenic stimuli in vitro. The secretome of Panc-MSCs was highly enriched for proteins associated with vascular development, wound healing and chemotaxis. Similar to BM-MSCs, Panc-MSCs conditioned media augmented endothelial cell survival, proliferation, and tubule formation in vitro. Importantly, the secretome of both MSC types was capable of stimulating chemotactic infiltration of murine endothelial cells in vivo and reduced hyperglycemia in STZ-treated mice following intrapancreatic injection. Overall, this study provides foundational knowledge to develop Panc-MSCs as a unique MSC subtype with functional properties beneficial in regenerative medicine for diabetes and vascular disease.
Collapse
Affiliation(s)
- Tyler T Cooper
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada.,Molecular Medicine Research Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute, London, Ontario, Canada.,Department of Biochemistry, Don Rix Protein Identification Facility, Western University, London, Ontario, Canada
| | - Stephen E Sherman
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada.,Molecular Medicine Research Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute, London, Ontario, Canada
| | - Gillian I Bell
- Molecular Medicine Research Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute, London, Ontario, Canada
| | - Jun Ma
- Molecular Medicine Research Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute, London, Ontario, Canada.,Department of Biochemistry, Don Rix Protein Identification Facility, Western University, London, Ontario, Canada
| | - Miljan Kuljanin
- Molecular Medicine Research Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute, London, Ontario, Canada.,Department of Biochemistry, Don Rix Protein Identification Facility, Western University, London, Ontario, Canada
| | - Shauna E Jose
- Molecular Medicine Research Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute, London, Ontario, Canada
| | - Gilles A Lajoie
- Department of Biochemistry, Don Rix Protein Identification Facility, Western University, London, Ontario, Canada
| | - David A Hess
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada.,Molecular Medicine Research Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute, London, Ontario, Canada
| |
Collapse
|
7
|
Mesenchymal Stromal Cell Therapy for Pancreatitis: A Systematic Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3250864. [PMID: 29743979 PMCID: PMC5878867 DOI: 10.1155/2018/3250864] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 12/31/2017] [Indexed: 12/19/2022]
Abstract
Background Based on animal studies, adult mesenchymal stromal cells (MSCs) are promising for the treatment of pancreatitis. However, the best type of this form of cell therapy and its mechanism of action remain unclear. Methods We searched the PubMed, Web of Science, Scopus, Google Scholar, and Clinical Trials.gov websites for studies using MSCs as a therapy for both acute and chronic pancreatitis published until September 2017. Results We identified 276 publications; of these publications, 18 met our inclusion criteria. In animal studies, stem cell therapy was applied more frequently for acute pancreatitis than for chronic pancreatitis. No clinical trials were identified. MSC therapy ameliorated pancreatic inflammation in acute pancreatitis and pancreatic fibrosis in chronic pancreatitis. Bone marrow and umbilical cord MSCs were the most frequently administered cell types. Due to the substantial heterogeneity among the studies regarding the type, source, and dose of MSCs used, conducting a meta-analysis was not feasible to determine the best type of MSCs. Conclusion The available data were insufficient for determining the best type of MSCs for the treatment of acute or chronic pancreatitis; therefore, clinical trials investigating the use of MSCs as therapy for pancreatitis are not warranted.
Collapse
|
8
|
Mathew E, Brannon AL, Del Vecchio A, Garcia PE, Penny MK, Kane KT, Vinta A, Buckanovich RJ, di Magliano MP. Mesenchymal Stem Cells Promote Pancreatic Tumor Growth by Inducing Alternative Polarization of Macrophages. Neoplasia 2016; 18:142-51. [PMID: 26992915 PMCID: PMC4796803 DOI: 10.1016/j.neo.2016.01.005] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 01/25/2016] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer is characterized by an extensive desmoplastic stroma, the functional relevance of which is poorly understood. Activated fibroblasts are a prevalent component of the stroma, and traditionally, these cells have been considered as a homogenous population derived from pancreatic stellate cells. In this study, we highlight a previously unappreciated heterogeneity of the fibroblast population within the stroma. In particular, a subset of stromal fibroblasts has characteristics of mesenchymal stem cells (MSCs). MSCs are present in the normal pancreas as well as in the carcinomatous pancreas (CA-MSCs). Here, we determine that CA-MSCs have increased tumor-promoting function compared with MSCs in normal pancreas. This ability to promote tumor growth is associated with CA-MSCs’ unique ability to promote alternative macrophage polarization. Thus, our study identifies a previously uncharacterized cell population within the stroma and sheds light on tumor-promoting interactions between different components of the stroma. Significance Targeting the stroma is emerging as a new paradigm in pancreatic cancer; however, efforts to that effect are hampered by our limited understanding of the nature and function of stromal components. Here, we uncover previously unappreciated heterogeneity within the stroma and identify interactions among stromal components that promote tumor growth and could be targeted therapeutically.
Collapse
Affiliation(s)
- Esha Mathew
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, 48109
| | - Arthur L Brannon
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, 48109; Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, 48109
| | | | - Paloma E Garcia
- Program in Pathology, University of Michigan, Ann Arbor, MI, 48109
| | - Morgan K Penny
- Program in Cancer Biology, University of Michigan, Ann Arbor, MI, 48109
| | - Kevin T Kane
- Department of Surgery, University of Michigan, Ann Arbor, MI, 48109
| | - Alekya Vinta
- Department of Surgery, University of Michigan, Ann Arbor, MI, 48109
| | - Ronald J Buckanovich
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, 48109; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109
| | - Marina Pasca di Magliano
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, 48109; Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, 48109; Department of Surgery, University of Michigan, Ann Arbor, MI, 48109; Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109.
| |
Collapse
|
9
|
Lee S, Jeong S, Lee C, Oh J, Kim SC. Mesenchymal Stem Cells Derived from Human Exocrine Pancreas Spontaneously Express Pancreas Progenitor-Cell Markers in a Cell-Passage-Dependent Manner. Stem Cells Int 2016; 2016:2142646. [PMID: 27630717 PMCID: PMC5007373 DOI: 10.1155/2016/2142646] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/21/2016] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cells (MSCs) derived from bone marrow, adipose tissue, and most connective tissues have been recognized as promising sources for cell-based therapies. MSCs have also been detected in human pancreatic tissue, including endocrine and exocrine cells. These adult human pancreas-derived MSCs have generated a great deal of interest owing to their potential use in the differentiation of insulin-producing cells for diabetes treatment. In the present study, we isolated MSCs from the adult human exocrine pancreas to determine whether isolated MSCs have the potential to differentiate into pancreatic endocrine cells and, therefore, whether they can be used in stem cell-based therapies. Pancreatic tissue was digested by collagenase and an enriched exocrine-cell fraction was obtained by density-gradient separation. Crude exocrine cells were methodically cultured in suspension and then in adherent culture. We expanded the human pancreatic exocrine-derived MSCs (hpMSCs) by cell passaging in culture and confirmed by flow cytometry that >90% expressed human classic surface markers of MSCs. Interestingly, these cells expressed pancreatic transcription factors, such as Pdx1, Ngn3, and MafA, similar to pancreatic progenitor cells. These results indicated that hpMSCs can be used for the differentiation of pancreatic endocrine cells and may be used in type 1 diabetes treatment.
Collapse
Affiliation(s)
- Song Lee
- Laboratory of Stem Cell Biology and Cell Therapy, Asan Institute for Life Science, Asan Medical Center, Seoul 05505, Republic of Korea
- Department of Surgery, University of Ulsan College of Medicine and Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Seonghee Jeong
- Laboratory of Stem Cell Biology and Cell Therapy, Asan Institute for Life Science, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Chanmi Lee
- Laboratory of Stem Cell Biology and Cell Therapy, Asan Institute for Life Science, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Jooyun Oh
- Laboratory of Stem Cell Biology and Cell Therapy, Asan Institute for Life Science, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Song-Cheol Kim
- Laboratory of Stem Cell Biology and Cell Therapy, Asan Institute for Life Science, Asan Medical Center, Seoul 05505, Republic of Korea
- Department of Surgery, University of Ulsan College of Medicine and Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| |
Collapse
|
10
|
Abstract
Since insulin discovery, islet transplantation was the first protocol to show the possibility to cure patients with type 1 diabetes using low-risk procedures. The scarcity of pancreas donors triggered a burst of studies focused on the production of new β cells in vitro. These were rapidly dominated by pluripotent stem cells (PSCs) demonstrating diabetes-reversal potential in diabetic mice. Subsequent enthusiasm fostered a clinical trial with immunoisolated embryonic-derived pancreatic progenitors. Yet safety is the Achilles' heel of PSCs, and a whole branch of β cell engineering medicine focuses on transdifferentiation of adult pancreatic cells. New data showed the possibility to chemically stimulate acinar or α cells to undergo β cell neogenesis and provide opportunities to intervene in situ without the need for a transplant, at least after weighing benefits against systemic adverse effects. The current studies suggested the pancreas as a reservoir of facultative progenitors (e.g., in the duct lining) could be exploited ex vivo for expansion and β cell differentiation in timely fashion and without the hurdles of PSC use. Diabetes cell therapy is thus a growing field not only with great potential but also with many pitfalls to overcome for becoming fully envisioned as a competitor to the current treatment standards.
Collapse
Affiliation(s)
- Philippe A Lysy
- Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium.
- Pediatric Endocrinology Unit, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium.
| | - Elisa Corritore
- Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium
| | - Etienne M Sokal
- Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
11
|
Laube M, Stolzing A, Thome UH, Fabian C. Therapeutic potential of mesenchymal stem cells for pulmonary complications associated with preterm birth. Int J Biochem Cell Biol 2016; 74:18-32. [PMID: 26928452 DOI: 10.1016/j.biocel.2016.02.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/23/2016] [Accepted: 02/25/2016] [Indexed: 12/22/2022]
Abstract
Preterm infants frequently suffer from pulmonary complications resulting in significant morbidity and mortality. Physiological and structural lung immaturity impairs perinatal lung transition to air breathing resulting in respiratory distress. Mechanical ventilation and oxygen supplementation ensure sufficient oxygen supply but enhance inflammatory processes which might lead to the establishment of a chronic lung disease called bronchopulmonary dysplasia (BPD). Current therapeutic options to prevent or treat BPD are limited and have salient side effects, highlighting the need for new therapeutic approaches. Mesenchymal stem cells (MSCs) have demonstrated therapeutic potential in animal models of BPD. This review focuses on MSC-based therapeutic approaches to treat pulmonary complications and critically compares results obtained in BPD models. Thereby bottlenecks in the translational systems are identified that are preventing progress in combating BPD. Notably, current animal models closely resemble the so-called "old" BPD with profound inflammation and injury, whereas clinical improvements shifted disease pathology towards a "new" BPD in which arrest of lung maturation predominates. Future studies need to evaluate the utility of MSC-based therapies in animal models resembling the "new" BPD though promising in vitro evidence suggests that MSCs do possess the potential to stimulate lung maturation. Furthermore, we address the mode-of-action of MSC-based therapies with regard to lung development and inflammation/fibrosis. Their therapeutic efficacy is mainly attributed to an enhancement of regeneration and immunomodulation due to paracrine effects. In addition, we discuss current improvement strategies by genetic modifications or precondition of MSCs to enhance their therapeutic efficacy which could also prove beneficial for BPD therapies.
Collapse
Affiliation(s)
- Mandy Laube
- Center for Pediatric Research Leipzig, Hospital for Children & Adolescents, Division of Neonatology, University of Leipzig, Leipzig, Germany.
| | - Alexandra Stolzing
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany; Loughborough University, Wolfson School of Mechanical and Manufacturing Engineering, Centre for Biological Engineering, Loughborough, UK.
| | - Ulrich H Thome
- Center for Pediatric Research Leipzig, Hospital for Children & Adolescents, Division of Neonatology, University of Leipzig, Leipzig, Germany.
| | - Claire Fabian
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany; Interdisciplinary Centre for Bioinformatics, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
12
|
Castinetti F, Brinkmeier ML, Mortensen AH, Vella KR, Gergics P, Brue T, Hollenberg AN, Gan L, Camper SA. ISL1 Is Necessary for Maximal Thyrotrope Response to Hypothyroidism. Mol Endocrinol 2015; 29:1510-21. [PMID: 26296153 DOI: 10.1210/me.2015-1192] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
ISLET1 is a homeodomain transcription factor necessary for development of the pituitary, retina, motor neurons, heart, and pancreas. Isl1-deficient mice (Isl1(-/-)) die early during embryogenesis at embryonic day 10.5 due to heart defects, and at that time, they have an undersized pituitary primordium. ISL1 is expressed in differentiating pituitary cells in early embryogenesis. Here, we report the cell-specific expression of ISL1 and assessment of its role in gonadotropes and thyrotropes. Isl1 expression is elevated in pituitaries of Cga(-/-) mice, a model of hypothyroidism with thyrotrope hypertrophy and hyperplasia. Thyrotrope-specific disruption of Isl1 with Tshb-cre is permissive for normal serum TSH, but T4 levels are decreased, suggesting decreased thyrotrope function. Inducing hypothyroidism in normal mice causes a reduction in T4 levels and dramatically elevated TSH response, but mice with thyrotrope-specific disruption of Isl1 have a blunted TSH response. In contrast, deletion of Isl1 in gonadotropes with an Lhb-cre transgene has no obvious effect on gonadotrope function or fertility. These results show that ISL1 is necessary for maximal thyrotrope response to hypothyroidism, in addition to its role in development of Rathke's pouch.
Collapse
Affiliation(s)
- F Castinetti
- Human Genetics, University of Michigan (F.C., M.L.B., A.H.M., P.G., S.A.C.), Ann Arbor, Michigan 48109; Beth Israel Deaconess Medical Center (K.R.V., A.N.H.), Harvard University, Boston, Massachusetts 02215; Aix-Marseille University (F.C., T.B.), Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Centre National de la Recherche Scientifique, Faculté de Médecine de Marseille, and Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Timone, Marseille, France 13000; and University of Rochester School of Medicine and Dentistry (L.G.), Rochester, New York 14642
| | - M L Brinkmeier
- Human Genetics, University of Michigan (F.C., M.L.B., A.H.M., P.G., S.A.C.), Ann Arbor, Michigan 48109; Beth Israel Deaconess Medical Center (K.R.V., A.N.H.), Harvard University, Boston, Massachusetts 02215; Aix-Marseille University (F.C., T.B.), Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Centre National de la Recherche Scientifique, Faculté de Médecine de Marseille, and Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Timone, Marseille, France 13000; and University of Rochester School of Medicine and Dentistry (L.G.), Rochester, New York 14642
| | - A H Mortensen
- Human Genetics, University of Michigan (F.C., M.L.B., A.H.M., P.G., S.A.C.), Ann Arbor, Michigan 48109; Beth Israel Deaconess Medical Center (K.R.V., A.N.H.), Harvard University, Boston, Massachusetts 02215; Aix-Marseille University (F.C., T.B.), Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Centre National de la Recherche Scientifique, Faculté de Médecine de Marseille, and Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Timone, Marseille, France 13000; and University of Rochester School of Medicine and Dentistry (L.G.), Rochester, New York 14642
| | - K R Vella
- Human Genetics, University of Michigan (F.C., M.L.B., A.H.M., P.G., S.A.C.), Ann Arbor, Michigan 48109; Beth Israel Deaconess Medical Center (K.R.V., A.N.H.), Harvard University, Boston, Massachusetts 02215; Aix-Marseille University (F.C., T.B.), Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Centre National de la Recherche Scientifique, Faculté de Médecine de Marseille, and Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Timone, Marseille, France 13000; and University of Rochester School of Medicine and Dentistry (L.G.), Rochester, New York 14642
| | - P Gergics
- Human Genetics, University of Michigan (F.C., M.L.B., A.H.M., P.G., S.A.C.), Ann Arbor, Michigan 48109; Beth Israel Deaconess Medical Center (K.R.V., A.N.H.), Harvard University, Boston, Massachusetts 02215; Aix-Marseille University (F.C., T.B.), Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Centre National de la Recherche Scientifique, Faculté de Médecine de Marseille, and Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Timone, Marseille, France 13000; and University of Rochester School of Medicine and Dentistry (L.G.), Rochester, New York 14642
| | - T Brue
- Human Genetics, University of Michigan (F.C., M.L.B., A.H.M., P.G., S.A.C.), Ann Arbor, Michigan 48109; Beth Israel Deaconess Medical Center (K.R.V., A.N.H.), Harvard University, Boston, Massachusetts 02215; Aix-Marseille University (F.C., T.B.), Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Centre National de la Recherche Scientifique, Faculté de Médecine de Marseille, and Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Timone, Marseille, France 13000; and University of Rochester School of Medicine and Dentistry (L.G.), Rochester, New York 14642
| | - A N Hollenberg
- Human Genetics, University of Michigan (F.C., M.L.B., A.H.M., P.G., S.A.C.), Ann Arbor, Michigan 48109; Beth Israel Deaconess Medical Center (K.R.V., A.N.H.), Harvard University, Boston, Massachusetts 02215; Aix-Marseille University (F.C., T.B.), Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Centre National de la Recherche Scientifique, Faculté de Médecine de Marseille, and Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Timone, Marseille, France 13000; and University of Rochester School of Medicine and Dentistry (L.G.), Rochester, New York 14642
| | - L Gan
- Human Genetics, University of Michigan (F.C., M.L.B., A.H.M., P.G., S.A.C.), Ann Arbor, Michigan 48109; Beth Israel Deaconess Medical Center (K.R.V., A.N.H.), Harvard University, Boston, Massachusetts 02215; Aix-Marseille University (F.C., T.B.), Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Centre National de la Recherche Scientifique, Faculté de Médecine de Marseille, and Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Timone, Marseille, France 13000; and University of Rochester School of Medicine and Dentistry (L.G.), Rochester, New York 14642
| | - S A Camper
- Human Genetics, University of Michigan (F.C., M.L.B., A.H.M., P.G., S.A.C.), Ann Arbor, Michigan 48109; Beth Israel Deaconess Medical Center (K.R.V., A.N.H.), Harvard University, Boston, Massachusetts 02215; Aix-Marseille University (F.C., T.B.), Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Centre National de la Recherche Scientifique, Faculté de Médecine de Marseille, and Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Timone, Marseille, France 13000; and University of Rochester School of Medicine and Dentistry (L.G.), Rochester, New York 14642
| |
Collapse
|
13
|
Giannoukakis N, Trucco M. Cellular therapies based on stem cells and their insulin-producing surrogates: a 2015 reality check. Pediatr Diabetes 2015; 16:151-63. [PMID: 25652322 DOI: 10.1111/pedi.12259] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 01/12/2015] [Indexed: 12/27/2022] Open
Abstract
Stem cell technology has recently gained a substantial amount of interest as one method to create a potentially limitless supply of transplantable insulin-producing cells to treat, and possibly cure diabetes mellitus. In this review, we summarize the state-of-the art of stem cell technology and list the potential sources of stem cells that have been shown to be useful as insulin-expressing surrogates. We also discuss the milestones that have been reached and those that remain to be addressed to generate bona fide beta cell-similar, insulin-producing surrogates. The caveats, limitations, and realistic expectations are also considered for current and future technology. In spite of the tremendous technical advances realized in the past decade, especially in the field of reprogramming adult somatic cells to become stem cells, the state-of-the art still relies on lengthy and cumbersome in vitro culture methods that yield cell populations that are not particularly glucose-responsive when transplanted into diabetic hosts. Despite the current impediments toward clinical translation, including the potential for immune rejection, the availability of technology to generate patient-specific reprogrammable stem cells has, and will be critical for, important insights into the genetics, epigenetics, biology, and physiology of insulin-producing cells in normal and pathologic states. This knowledge could accelerate the time to reach the desired breakthrough for safe and efficacious beta cell surrogates.
Collapse
Affiliation(s)
- Nick Giannoukakis
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA, USA
| | | |
Collapse
|
14
|
Abstract
Diabetes is a common multisystem disease that results in hyperglycemia due to a relative or absolute insulin deficiency. Improved glycemic control decreases the risk of development and progression of microvascular and, to a lesser extent, macrovascular complications and prevents symptomatic hyperglycemia. However, complex treatment regimens aimed at improving glycemic control are associated with an increased incidence of hypoglycemia. On paper at least, cellular therapies arising from reprogramed stem cells or other somatic cell types would provide ideal therapy for diabetes and the prevention of its complications. This hypothesis has led to intensive efforts to grow β cells from various sources. In this review, we provide an overview of β-cell development as well as the efforts reported to date in terms of cellular therapy for diabetes. Engineering β-cell replacement therapy requires an understanding of how β cells respond to other metabolites such as amino acids, free fatty acids, and ketones. Indeed, efforts thus far have been characterized by an inability of cellular replacement products to adequately respond to metabolites that normally couple the metabolic state to β-cell function and insulin secretion. Efforts to date intended to capitalize on current knowledge of islet cell development and stimulus-secretion coupling of the β cell are encouraging but as yet of little clinical relevance.
Collapse
Affiliation(s)
- Aleksey Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| | - Adrian Vella
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN.
| |
Collapse
|
15
|
Lejmi E, Perriraz N, Clément S, Morel P, Baertschiger R, Christofilopoulos P, Meier R, Bosco D, Bühler LH, Gonelle-Gispert C. Inflammatory Chemokines MIP-1δ and MIP-3α Are Involved in the Migration of Multipotent Mesenchymal Stromal Cells Induced by Hepatoma Cells. Stem Cells Dev 2015; 24:1223-35. [PMID: 25579056 DOI: 10.1089/scd.2014.0176] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In vivo, bone marrow-derived multipotent mesenchymal stromal cells (MSC) have been identified at sites of tumors, suggesting that specific signals mobilize and activate MSC to migrate to areas surrounding tumors. The signals and migratory mechanisms that guide MSC are not well understood. Here, we investigated the migration of human MSC induced by conditioned medium of Huh-7 hepatoma cells (Huh-7 CM). Using a transwell migration system, we showed that human MSC migration was increased in the presence of Huh-7 CM. Using a human cytokine antibody array, we detected increased levels of MIP-1δ and MIP-3α in Huh-7 CM. Recombinant chemokines MIP-1δ and MIP-3α induced MSC migration. Anti-MIP-1δ and anti-MIP-3α antibodies added to Huh-7 CM decreased MSC migration, further suggesting that MIP-1δ and MIP-3α were implicated in the Huh-7 CM-induced MSC migration. By real-time polymerase chain reaction, we observed an absence of chemokine receptors CCR2 and CXCR2 and low expression of CCR1, CCR5, and CCR6 in MSC. Expression of these chemokine receptors was not regulated by Huh-7 CM. Furthermore, matrix metalloproteinase 1 (MMP-1) expression was strongly increased in MSC after incubation with Huh-7 CM, suggesting that MSC migration depends on MMP-1 activity. The signaling pathway MAPK/ERK was activated by Huh-7 CM but its inhibition by PD98059 did not impair Huh-7 CM-induced MSC migration. Further, long-term incubation of MSC with MIP-1δ increased α-smooth muscle actin expression, suggesting its implication in the Huh-7 CM-induced evolvement of MSC into myofibroblasts. In conclusion, we report that two inflammatory cytokines, MIP-1δ and MIP-3α, are able to increase MSC migration in vitro. These cytokines might be responsible for migration and evolvement of MSC into myofibroblasts around tumors.
Collapse
Affiliation(s)
- Esma Lejmi
- 1 Surgical Research Unit, University Hospitals of Geneva , Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Meier RPH, Mahou R, Morel P, Meyer J, Montanari E, Muller YD, Christofilopoulos P, Wandrey C, Gonelle-Gispert C, Bühler LH. Microencapsulated human mesenchymal stem cells decrease liver fibrosis in mice. J Hepatol 2015; 62:634-641. [PMID: 25450712 DOI: 10.1016/j.jhep.2014.10.030] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 10/15/2014] [Accepted: 10/20/2014] [Indexed: 01/07/2023]
Abstract
BACKGROUND & AIMS Mesenchymal stem cell (MSC) transplantation was shown to be effective for the treatment of liver fibrosis, but the mechanisms of action are not yet fully understood. We transplanted encapsulated human MSCs in two mouse models of liver fibrosis to determine the mechanisms behind the protective effect. METHODS Human bone marrow-derived MSCs were microencapsulated in novel alginate-polyethylene glycol microspheres. In vitro, we analyzed the effect of MSC-conditioned medium on the activation of hepatic stellate cells and the viability, proliferation, cytokine secretion, and differentiation capacity of encapsulated MSCs. The level of fibrosis induced by bile duct ligation (BDL) or carbon tetrachloride (CCl4) was assessed after intraperitoneal transplantation of encapsulated MSCs, encapsulated human fibroblasts, and empty microspheres. RESULTS MSC-conditioned medium inhibited hepatic stellate cell activation and release of MSC secreted anti-apoptotic (IL-6, IGFBP-2) and anti-inflammatory (IL-1Ra) cytokines. Viability, proliferation, and cytokine secretion of microencapsulated MSCs were similar to those of non-encapsulated MSCs. Within the microspheres, MSCs maintained their capacity to differentiate into adipocytes, chondrocytes, and osteocytes. 23% (5/22) of the MSC clones were able to produce anti-inflammatory IL-1Ra in vitro. Microencapsulated MSCs significantly delayed the development of BDL- and CCl4-induced liver fibrosis. Fibroblasts had an intermediate effect against CCl4-induced fibrosis. Mice transplanted with encapsulated MSCs showed lower mRNA levels of collagen type I, whereas levels of matrix metalloproteinase 9 were significantly higher. Human IL-1Ra was detected in the serum of 36% (4/11) of the mice transplanted with microencapsulated MSCs. CONCLUSIONS MSC-derived soluble molecules are responsible for an anti-fibrotic effect in experimental liver fibrosis.
Collapse
Affiliation(s)
- Raphael P H Meier
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and Medical School, Geneva, Switzerland.
| | - Redouan Mahou
- Institut d'Ingénierie Biologique et Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Philippe Morel
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and Medical School, Geneva, Switzerland
| | - Jeremy Meyer
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and Medical School, Geneva, Switzerland
| | - Elisa Montanari
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and Medical School, Geneva, Switzerland
| | - Yannick D Muller
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and Medical School, Geneva, Switzerland
| | | | - Christine Wandrey
- Institut d'Ingénierie Biologique et Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Carmen Gonelle-Gispert
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and Medical School, Geneva, Switzerland
| | - Leo H Bühler
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and Medical School, Geneva, Switzerland
| |
Collapse
|
17
|
Fokas E, O'Neill E, Gordon-Weeks A, Mukherjee S, McKenna WG, Muschel RJ. Pancreatic ductal adenocarcinoma: From genetics to biology to radiobiology to oncoimmunology and all the way back to the clinic. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1855:61-82. [PMID: 25489989 DOI: 10.1016/j.bbcan.2014.12.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Revised: 12/01/2014] [Accepted: 12/03/2014] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer death. Despite improvements in the clinical management, the prognosis of PDAC remains dismal. In the present comprehensive review, we will examine the knowledge of PDAC genetics and the new insights into human genome sequencing and clonal evolution. Additionally, the biology and the role of the stroma in tumour progression and response to treatment will be presented. Furthermore, we will describe the evidence on tumour chemoresistance and radioresistance and will provide an overview on the recent advances in PDAC metabolism and circulating tumour cells. Next, we will explore the characteristics and merits of the different mouse models of PDAC. The inflammatory milieu and the immunosuppressive microenvironment mediate tumour initiation and treatment failure. Hence, we will also review the inflammatory and immune escaping mechanisms and the new immunotherapies tested in PDAC. A better understanding of the different mechanisms of tumour formation and progression will help us to identify the best targets for testing in future clinical studies of PDAC.
Collapse
MESH Headings
- Animals
- Cancer Vaccines/therapeutic use
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/therapy
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/pathology
- Disease Models, Animal
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/immunology
- Humans
- Immunotherapy/methods
- Inflammation/pathology
- Mice
- Neoplastic Cells, Circulating/immunology
- Neoplastic Cells, Circulating/pathology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/therapy
- Radiation Tolerance/genetics
Collapse
Affiliation(s)
- Emmanouil Fokas
- Department of Oncology, Oxford Institute for Radiation Oncology, Oxford University, Oxford, UK.
| | - Eric O'Neill
- Department of Oncology, Oxford Institute for Radiation Oncology, Oxford University, Oxford, UK
| | - Alex Gordon-Weeks
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Somnath Mukherjee
- Department of Oncology, Oxford Institute for Radiation Oncology, Oxford University, Oxford, UK
| | - W Gillies McKenna
- Department of Oncology, Oxford Institute for Radiation Oncology, Oxford University, Oxford, UK
| | - Ruth J Muschel
- Department of Oncology, Oxford Institute for Radiation Oncology, Oxford University, Oxford, UK
| |
Collapse
|
18
|
Orlando G, Gianello P, Salvatori M, Stratta RJ, Soker S, Ricordi C, Domínguez-Bendala J. Cell replacement strategies aimed at reconstitution of the β-cell compartment in type 1 diabetes. Diabetes 2014; 63:1433-44. [PMID: 24757193 DOI: 10.2337/db13-1742] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Emerging technologies in regenerative medicine have the potential to restore the β-cell compartment in diabetic patients, thereby overcoming the inadequacies of current treatment strategies and organ supply. Novel approaches include: 1) Encapsulation technology that protects islet transplants from host immune surveillance; 2) stem cell therapies and cellular reprogramming, which seek to regenerate the depleted β-cell compartment; and 3) whole-organ bioengineering, which capitalizes on the innate properties of the pancreas extracellular matrix to drive cellular repopulation. Collaborative efforts across these subfields of regenerative medicine seek to ultimately produce a bioengineered pancreas capable of restoring endocrine function in patients with insulin-dependent diabetes.
Collapse
|
19
|
Wang Y, Liu J, Tan X, Li G, Gao Y, Liu X, Zhang L, Li Y. Induced pluripotent stem cells from human hair follicle mesenchymal stem cells. Stem Cell Rev Rep 2014; 9:451-60. [PMID: 23242965 PMCID: PMC3742959 DOI: 10.1007/s12015-012-9420-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Reprogramming of somatic cells into inducible pluripotent stem cells (iPSCs) provides an alternative to using embryonic stem cells (ESCs). Mesenchymal stem cells derived from human hair follicles (hHF-MSCs) are easily accessible, reproducible by direct plucking of human hairs. Whether these hHF-MSCs can be reprogrammed has not been previously reported. Here we report the generation of iPSCs from hHF-MSCs obtained by plucking several hairs. hHF-MSCs were isolated from hair follicle tissues and their mesenchymal nature confirmed by detecting cell surface antigens and multilineage differentiation potential towards adipocytes and osteoblasts. They were then reprogrammed into iPSCs by lentiviral transduction with Oct4, Sox2, c-Myc and Klf4. hHF-MSC-derived iPSCs appeared indistinguishable from human embryonic stem cells (hESCs) in colony morphology, expression of alkaline phosphotase, and expression of specific hESCs surface markers, SSEA-3, SSEA-4, Tra-1-60, Tra-1-81, Nanog, Oct4, E-Cadherin and endogenous pluripotent genes. When injected into immunocompromised mice, hHF-MSC-derived iPSCs formed teratomas containing representatives of all three germ layers. This is the first study to report reprogramming of hHF-MSCs into iPSCs.
Collapse
Affiliation(s)
- Yimei Wang
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun 130021, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Cerf ME. Beta cell dynamics: beta cell replenishment, beta cell compensation and diabetes. Endocrine 2013; 44:303-11. [PMID: 23483434 DOI: 10.1007/s12020-013-9917-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 03/01/2013] [Indexed: 12/19/2022]
Abstract
Type 2 diabetes, characterized by persistent hyperglycemia, arises mostly from beta cell dysfunction and insulin resistance and remains a highly complex metabolic disease due to various stages in its pathogenesis. Glucose homeostasis is primarily regulated by insulin secretion from the beta cells in response to prevailing glycemia. Beta cell populations are dynamic as they respond to fluctuating insulin demand. Beta cell replenishment and death primarily regulate beta cell populations. Beta cells, pancreatic cells, and extra-pancreatic cells represent the three tiers for replenishing beta cells. In rodents, beta cell self-replenishment appears to be the dominant source for new beta cells supported by pancreatic cells (non-beta islet cells, acinar cells, and duct cells) and extra-pancreatic cells (liver, neural, and stem/progenitor cells). In humans, beta cell neogenesis from non-beta cells appears to be the dominant source of beta cell replenishment as limited beta cell self-replenishment occurs particularly in adulthood. Metabolic states of increased insulin demand trigger increased insulin synthesis and secretion from beta cells. Beta cells, therefore, adapt to support their physiology. Maintaining physiological beta cell populations is a strategy for targeting metabolic states of persistently increased insulin demand as in diabetes.
Collapse
Affiliation(s)
- Marlon E Cerf
- Diabetes Discovery Platform, South African Medical Research, PO Box 19070, Tygerberg, 7505, South Africa,
| |
Collapse
|
21
|
Abstract
Cell therapy is currently considered as a potential therapeutic alternative to traditional treatments of diabetes. Islet and whole pancreas transplantations provided the proof-of-concept of glucose homeostasis restoration after replenishment of the deficiency of β cells responsible for the disease. Current limitations of these procedures have led to the search for strategies targeting replication of pre-existing β cells or transdifferentiation of progenitors and adult cells. These investigations revealed an unexpected plasticity towards β cells of adult cells residing in pancreatic epithelium (eg, acinar, duct, and α cells). Here we discuss recent developments in β-cell replication and β-cell transdifferentiation of adult epithelial pancreatic cells, with an emphasis on techniques with a potential for clinical translation.
Collapse
Affiliation(s)
| | | | - Susan Bonner-Weir
- Correspondence to: Susan Bonner-Weir, PhD, Joslin Diabetes Center, 1 Joslin Place, Boston, MA 02215, USA; ; Phone 617-309-2581, Fax 617-309-2650
| |
Collapse
|
22
|
Transplantation of mesenchymal stem cells for the treatment of liver diseases, is there enough evidence? Stem Cell Res 2013; 11:1348-64. [PMID: 24090934 DOI: 10.1016/j.scr.2013.08.011] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 08/18/2013] [Accepted: 08/21/2013] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells or multipotent mesenchymal stromal cells (MSCs) have been extensively investigated in small animal models to treat both acute and chronic liver injuries. Mechanisms of action are not clearly elucidated but may include their ability to differentiate into hepatocyte-like cells, to reduce inflammation, and to enhance tissue repair at the site of injury. This approach is controversial and evidence in large animals is missing. Side effects of MSC infusion such as the contribution to a fibrotic process have been reported in experimental settings. Nevertheless, MSCs moved quickly from bench to bedside and over 280 clinical trials are registered, of which 28 focus on the treatment of liver diseases. If no severe side-effects were observed so far, long-term benefits remain uncertain. More preclinical data regarding mechanisms of action, long term safety and efficacy are warranted before initiating large scale clinical application. The proposal of this review is to visit the current state of knowledge regarding mechanisms behind the therapeutic effects of MSCs in the treatment of experimental liver diseases, to address questions about efficacy and risk, and to discuss recent clinical advances involving MSC-based therapies.
Collapse
|
23
|
Hematti P, Kim J, Stein AP, Kaufman D. Potential role of mesenchymal stromal cells in pancreatic islet transplantation. Transplant Rev (Orlando) 2013; 27:21-9. [PMID: 23290684 DOI: 10.1016/j.trre.2012.11.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 11/09/2012] [Indexed: 12/11/2022]
Abstract
Pancreatic islet transplantation is an attractive option for treatment of type 1 diabetes mellitus but maintaining long term islet function remains challenging. Mesenchymal stromal cells (MSCs), derived from bone marrow or other sources, are being extensively investigated in the clinical setting for their immunomodulatory and tissue regenerative properties. Indeed, MSCs have been already tested in some feasibility studies in the context of islet transplantation. MSCs could be utilized to improve engraftment of pancreatic islets by suppressing inflammatory damage and immune mediated rejection. In addition to their immunomodulatory effects, MSCs are known to provide a supportive microenvironmental niche by secreting paracrine factors and depositing extracellular matrix. These properties could be used for in vivo co-transplantation to improve islet engraftment, or for in vitro co-culture to prime freshly isolated islets prior to implantation. Further, tissue specific pancreatic islet derived MSCs may open new opportunities for its use in islet transplantation as those cells might be more physiological to pancreatic islets.
Collapse
Affiliation(s)
- Peiman Hematti
- Department of Medicine, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA.
| | | | | | | |
Collapse
|
24
|
Domínguez-Bendala J, Ricordi C. Present and future cell therapies for pancreatic beta cell replenishment. World J Gastroenterol 2012; 18:6876-84. [PMID: 23322984 PMCID: PMC3531670 DOI: 10.3748/wjg.v18.i47.6876] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 05/27/2012] [Accepted: 07/18/2012] [Indexed: 02/06/2023] Open
Abstract
If only at a small scale, islet transplantation has successfully addressed what ought to be the primary endpoint of any cell therapy: the functional replenishment of damaged tissue in patients. After years of less-than-optimal approaches to immunosuppression, recent advances consistently yield long-term graft survival rates comparable to those of whole pancreas transplantation. Limited organ availability is the main hurdle that stands in the way of the widespread clinical utilization of this pioneering intervention. Progress in stem cell research over the past decade, coupled with our decades-long experience with islet transplantation, is shaping the future of cell therapies for the treatment of diabetes. Here we review the most promising avenues of research aimed at generating an inexhaustible supply of insulin-producing cells for islet regeneration, including the differentiation of pluripotent and multipotent stem cells of embryonic and adult origin along the beta cell lineage and the direct reprogramming of non-endocrine tissues into insulin-producing cells.
Collapse
|
25
|
Ricordi C, Inverardi L, Domínguez-Bendala J. From cellular therapies to tissue reprogramming and regenerative strategies in the treatment of diabetes. Regen Med 2012; 7:41-8. [DOI: 10.2217/rme.12.70] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Diabetes mellitus represents a global epidemic affecting over 350 million patients worldwide and projected by the WHO to surpass the 500 million patient mark within the next two decades. Besides Type 1 and Type 2 diabetes mellitus, the study of the endocrine compartment of the pancreas is of great translational interest, as strategies aimed at restoring its mass could become therapies for glycemic dysregulation, drug-related diabetes following diabetogenic therapies, or hyperglycemic disturbances following the treatment of cancer and nesidioblastosis. Such strategies generally fall under one of the ‘three Rs’: replacement (islet transplantation and stem cell differentiation); reprogramming (e.g., from the exocrine compartment of the pancreas); and regeneration (replication and induction of endogenous stem cells). As the latter has been extensively reviewed in recent months by us and others, this article focuses on emerging reprogramming and replacement approaches.
Collapse
Affiliation(s)
- Camillo Ricordi
- University of Miami Cell Transplant Center and Diabetes Research Institute, Miami, FL, USA
| | - Luca Inverardi
- University of Miami Cell Transplant Center and Diabetes Research Institute, Miami, FL, USA
| | - Juan Domínguez-Bendala
- University of Miami Cell Transplant Center and Diabetes Research Institute, Miami, FL, USA
| |
Collapse
|
26
|
Characterization of myelomonocytoid progenitor cells with mesenchymal differentiation potential obtained by outgrowth from pancreas explants. BIOTECHNOLOGY RESEARCH INTERNATIONAL 2012; 2012:429868. [PMID: 22953065 PMCID: PMC3431127 DOI: 10.1155/2012/429868] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Revised: 06/06/2012] [Accepted: 06/14/2012] [Indexed: 01/16/2023]
Abstract
Progenitor cells can be obtained by outgrowth from tissue explants during primary ex vivo tissue culture. We have isolated and characterized cells outgrown from neonatal mouse pancreatic explants. A relatively uniform population of cells showing a distinctive morphology emerged over time in culture. This population expressed monocyte/macrophage and hematopoietic markers (CD11b(+) and CD45(+)), and some stromal-related markers (CD44(+) and CD29(+)), but not mesenchymal stem cell (MSC)-defining markers (CD90(-) and CD105(-)) nor endothelial (CD31(-)) or stem cell-associated markers (CD133(-) and stem cell antigen-1; Sca-1(-)). Cells could be maintained in culture as a plastic-adherent monolayer in culture medium (MesenCult MSC) for more than 1 year. Cells spontaneously formed sphere clusters "pancreatospheres" which, however, were nonclonal. When cultured in appropriate media, cells differentiated into multiple mesenchymal lineages (fat, cartilage, and bone). Positive dithizone staining suggested that a subset of cells differentiated into insulin-producing cells. However, further studies are needed to characterize the endocrine potential of these cells. These findings indicate that a myelomonocytoid population from pancreatic explant outgrowths has mesenchymal differentiation potential. These results are in line with recent data onmonocyte-derivedmesenchymal progenitors (MOMPs).
Collapse
|
27
|
Adipose derived mesenchymal stem cells transplantation via portal vein improves microcirculation and ameliorates liver fibrosis induced by CCl4 in rats. J Transl Med 2012; 10:133. [PMID: 22735033 PMCID: PMC3439354 DOI: 10.1186/1479-5876-10-133] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 06/19/2012] [Indexed: 02/08/2023] Open
Abstract
Introduction Adipose derived mesenchymal stem cells (ADMSCs), carrying the similar characteristics to bone marrow mesenchymal stem cells, only much more abundant and easier to obtain, may be a promising treatment for liver fibrosis. We aim to investigate the therapeutic potential of ADMSCs transplantation in liver fibrosis caused by carbon tetrachloride (CCl4) in rats as well as its underlying mechanism, and to further explore the appropriate infusion pathway. Methods ADMSCs were isolated, cultured and identified. Placebo and ADMSCs were transplanted via portal vein and tail vein respectively into carbon tetrachloride (CCl4)-induced liver fibrosis rats. Computed tomography (CT) perfusion scan and microvessel counts were performed to measure the alteration of liver microcirculation after therapy. Liver function tests and histological findings were estimated. Results CT perfusion scan shown significant decrease of hepatic arterial perfusion index, significant increased portal vein perfusion, total liver perfusion in rats receiving ADMSCs from portal vein, and Factor VIII (FVIII) immunohistochemical staining shown significant decrease of microvessels in rats receiving ADMSCs from portal vein, indicating microcirculation improvement in portal vein group. Vascular endothelial growth Factor (VEGF) was significantly up-regulated in fibrosis models, and decreased after ADMSCs intraportal transplantation. A significant improvement of liver functional test and histological findings in portal vein group were observed. No significance was found in rats receiving ADMSCs from tail vein. Conclusions ADMSCs have a therapeutic effect against CCl4-mediated liver fibrosis. ADMSCs may benefit the fibrotic liver through alteration of microcirculation, evidenced by CT perfusion scan and down-regulation of VEGF. Intraportal transplantation is a better pathway than tail vein transplantation.
Collapse
|
28
|
Lysy PA, Weir GC, Bonner-Weir S. Concise review: pancreas regeneration: recent advances and perspectives. Stem Cells Transl Med 2012. [PMID: 23197762 DOI: 10.5966/sctm.2011-0025] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The replacement of functional pancreatic β-cells is seen as an attractive potential therapy for diabetes, because diabetes results from an inadequate β-cell mass. Inducing replication of the remaining β-cells and new islet formation from progenitors within the pancreas (neogenesis) are the most direct ways to increase the β-cell mass. Stimulation of both replication and neogenesis have been reported in rodents, but their clinical significance must still be shown. Because human islet transplantation is limited by the scarcity of donors and graft failure within a few years, efforts have recently concentrated on the use of stem cells to replace the deficient β-cells. Currently, embryonic stem cells and induced pluripotent stem cells achieve high levels of β-cell differentiation, but their clinical use is still hampered by ethical issues and/or the risk of developing tumors after transplantation. Pancreatic epithelial cells (duct, acinar, or α-cells) represent an appealing alternative to stem cells because they demonstrate β-cell differentiation capacities. Yet translation of such capacity to human cells after significant in vitro expansion has yet to be achieved. Besides providing new β-cells, cell therapy also has to address the question on how to protect the transplanted cells from destruction by the immune system via either allo- or autoimmunity. Encouraging developments have been made in encapsulation and immunomodulation techniques, but many challenges still remain. Herein, we discuss recent advances in the search for β-cell replacement therapies, current strategies for circumventing the immune system, and mandatory steps for new techniques to be translated from bench to clinics.
Collapse
Affiliation(s)
- Philippe A Lysy
- Joslin Diabetes Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
29
|
Prabakar KR, Domínguez-Bendala J, Molano RD, Pileggi A, Villate S, Ricordi C, Inverardi L. Generation of glucose-responsive, insulin-producing cells from human umbilical cord blood-derived mesenchymal stem cells. Cell Transplant 2011; 21:1321-39. [PMID: 22195604 DOI: 10.3727/096368911x612530] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
We sought to assess the potential of human cord blood-derived mesenchymal stem cells (CB-MSCs) to derive insulin-producing, glucose-responsive cells. We show here that differentiation protocols based on stepwise culture conditions initially described for human embryonic stem cells (hESCs) lead to differentiation of cord blood-derived precursors towards a pancreatic endocrine phenotype, as assessed by marker expression and in vitro glucose-regulated insulin secretion. Transplantation of these cells in immune-deficient animals shows human C-peptide production in response to a glucose challenge. These data suggest that human cord blood may be a promising source for regenerative medicine approaches for the treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Kamalaveni R Prabakar
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Domínguez-Bendala J, Lanzoni G, Inverardi L, Ricordi C. Concise review: mesenchymal stem cells for diabetes. Stem Cells Transl Med 2011. [PMID: 23197641 DOI: 10.5966/sctm.2011-0017] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have already made their mark in the young field of regenerative medicine. Easily derived from many adult tissues, their therapeutic worth has already been validated for a number of conditions. Unlike embryonic stem cells, neither their procurement nor their use is deemed controversial. Here we review the potential use of MSCs for the treatment of type 1 diabetes mellitus, a devastating chronic disease in which the insulin-producing cells of the pancreas (the β-cells) are the target of an autoimmune process. It has been hypothesized that stem cell-derived β-cells may be used to replenish the islet mass in diabetic patients, making islet transplantation (a form of cell therapy that has already proven effective at clinically restoring normoglycemia) available to millions of prospective patients. Here we review the most current advances in the design and application of protocols for the differentiation of transplantable β-cells, with a special emphasis in analyzing MSC potency according to their tissue of origin. Although no single method appears to be ripe enough for clinical trials yet, recent progress in reprogramming (a biotechnological breakthrough that relativizes the thus far insurmountable barriers between embryonal germ layers) bodes well for the rise of MSCs as a potential weapon of choice to develop personalized therapies for type 1 diabetes.
Collapse
|
31
|
β-Cell Generation: Can Rodent Studies Be Translated to Humans? J Transplant 2011; 2011:892453. [PMID: 22007286 PMCID: PMC3189575 DOI: 10.1155/2011/892453] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 07/31/2011] [Accepted: 07/31/2011] [Indexed: 12/26/2022] Open
Abstract
β-cell replacement by allogeneic islet transplantation is a promising approach for patients with type 1 diabetes, but the shortage of organ donors requires new sources of β cells. Islet regeneration in vivo and generation of β-cells ex vivo followed by transplantation represent attractive therapeutic alternatives to restore the β-cell mass. In this paper, we discuss different postnatal cell types that have been envisaged as potential sources for future β-cell replacement therapy. The ultimate goal being translation to the clinic, a particular attention is given to the discrepancies between findings from studies performed in rodents (both ex vivo on primary cells and in vivo on animal models), when compared with clinical data and studies performed on human cells.
Collapse
|
32
|
Sumi S. Regenerative medicine for insulin deficiency: creation of pancreatic islets and bioartificial pancreas. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2011; 18:6-12. [PMID: 20589399 DOI: 10.1007/s00534-010-0303-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Recent advances in pancreas organogenesis have greatly improved the understanding of cell lineage from inner cell mass to fully differentiated β-cells. Based upon such knowledge, insulin-producing cells similar to β-cells to a certain extent have been generated from various cell sources including embryonic stem cells (ESCs) and induced pluripotent stem (iPS) cells, although fully differentiated cells comparable to β-cells are not yet available. The bioartificial pancreas is a therapeutic approach to enable allo- and xenotransplantation of islets without immune suppression. Among several types of bioartificial pancreases (BAPs), micro-encapsulated porcine islets are already in use in clinical trials and may, perhaps, replace islet transplantation in the near future. Some types of bioartificial pancreas such as macro-encapsulation are also useful for keeping transplanted cells enclosed in case retrieval is necessary. Therefore, early clinical applications of artificially generated β-like cells, especially those from ESCs or iPS cells, will be considered in combination with retrievable BAPs.
Collapse
Affiliation(s)
- Shoichiro Sumi
- Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW The promise of islet transplantation for type 1 diabetes has been hampered by the lack of a renewable source of insulin-producing cells. However, steadfast advances in the field have set the stage for stem cell-based approaches to take over in the near future. This review focuses on the most intriguing findings reported in recent years, which include not only progress in adult and embryonic stem cell differentiation, but also the direct reprogramming of nonendocrine tissues into insulin-producing beta cells. RECENT FINDINGS In spite of their potential for tumorigenesis, human embryonic stem (hES) cells are poised to be in clinical trials within the next decade. This situation is mainly due to the preclinical success of a differentiation method that recapitulates beta cell development. In contrast, adult stem cells still need one such gold standard of differentiation, and progress is somewhat impeded by the lack of consensus on the best source. A concerted effort is necessary to bring their potential to clinical fruition. In the meantime, reported success in reprogramming might offer a 'third way' towards the rescue of pancreatic endocrine function. SUMMARY Here we discuss the important strategic decisions that need to be made in order to maximize the therapeutic chances of each of the presented approaches.
Collapse
Affiliation(s)
- Juan Domínguez-Bendala
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine; 1450 NW 10 Ave, Miami, FL 33136
- Department of Surgery, University of Miami Miller School of Medicine
| | - Luca Inverardi
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine; 1450 NW 10 Ave, Miami, FL 33136
- Department of Medicine, University of Miami Miller School of Medicine
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine; 1450 NW 10 Ave, Miami, FL 33136
- Department of Surgery, University of Miami Miller School of Medicine
| |
Collapse
|
34
|
Seeberger KL, Eshpeter A, Korbutt GS. Isolation and culture of human multipotent stromal cells from the pancreas. Methods Mol Biol 2011; 698:123-140. [PMID: 21431515 DOI: 10.1007/978-1-60761-999-4_10] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Mesenchymal stem cells, also termed multipotent mesenchymal stromal cells (MSCs), can be isolated from most adult tissues. Although the exact origin of MSCs expanded from the human pancreas has not been resolved, we have developed protocols to isolate and expand MSCs from human pancreatic tissue that remains after islet procurement. Similar to techniques used to isolate MSCs from bone marrow, pancreatic MSCs are isolated based on their cell adherence, expression of several cell surface antigens, and multilineage differentiation. The protocols for isolating, characterizing, and differentiating MSCs from the pancreas are presented in this chapter.
Collapse
Affiliation(s)
- Karen L Seeberger
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | | | | |
Collapse
|
35
|
Fanjul M, Gmyr V, Sengenès C, Ratovo G, Dufresne M, Lefebvre B, Kerr-Conte J, Hollande E. Evidence for epithelial-mesenchymal transition in adult human pancreatic exocrine cells. J Histochem Cytochem 2010; 58:807-23. [PMID: 20530463 DOI: 10.1369/jhc.2010.955807] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
It has been shown that adult pancreatic ductal cells can dedifferentiate and act as pancreatic progenitors. Dedifferentiation of epithelial cells is often associated with the epithelial-mesenchymal transition (EMT). In this study, we investigated the occurrence of EMT in adult human exocrine pancreatic cells both in vitro and in vivo. Cells of exocrine fraction isolated from the pancreas of brain-dead donors were first cultured in suspension for eight days. This led to the formation of spheroids, composed of a principal population of cells with duct-like phenotype. When cultivated in tissue culture-treated flasks, spheroid cells exhibited a proliferative capacity and coexpressed epithelial (cytokeratin7 and cytokeratin19) and mesenchymal (vimentin and alpha-smooth muscle actin) markers as well as marker of progenitor pancreatic cells (pancreatic duodenal homeobox factor-1) and surface markers of mesenchymal stem cells. The switch from E-cadherin to N-cadherin associated with Snail1 expression suggested that these cells underwent EMT. In addition, we showed coexpression of epithelial and mesenchymal markers in ductal cells of one normal adult pancreas and three type 2 diabetic pancreases. Some of the vimentin-positive cells were found to coexpress glucagon or amylase. These results point to the occurrence of EMT, which may take place on dedifferentiation of ductal cells during the regeneration or renewal of human pancreatic tissues.
Collapse
Affiliation(s)
- Marjorie Fanjul
- Institut National de la Santé et de la Recherche Médicale U858, Toulouse, France
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Levetan C. Distinctions between islet neogenesis and β-cell replication: implications for reversal of Type 1 and 2 diabetes. J Diabetes 2010; 2:76-84. [PMID: 20923488 DOI: 10.1111/j.1753-0407.2010.00074.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The terms "islet" and "β-cell" are often used interchangeably, yet islets are highly complex multicellular organelles that contain the insulin-producing β-cells and four other cells types, all of which play a role in maintaining glucose homeostasis within a very narrow range. Although the formation of new islets in adults is rare, occurring primarily in response to pancreatic injury and major stress to the pancreas, β-cell replication from existing cells occurs throughout adulthood. An understanding of the regulatory factors controlling pancreatic development has more clearly defined the differences between new islet formation from progenitor cells located throughout the adult pancreas and β-cell replication occurring within existing islets. The present review sets forth to more clearly distinguish the differences between the postnatal pathways of islet neogenesis and β-cell replication with a discussion of the potential implications for reversal of Type 1 and 2 diabetic patients using islet neogenesis agents that are now in development. For Type 1 diabetic patients, an immune tolerance agent in conjunction with an islet neogenesis agent may allow achievement of adequate islet mass, perhaps with subsequent potential to withdraw medications. For Type 2 diabetic patients, lifestyle changes and/or medications may sustain the production of new islets and limit the accelerated β-cell apoptosis characteristic of the condition.
Collapse
Affiliation(s)
- Claresa Levetan
- Division of Endocrinology, Chestnut Hill Hospital, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
37
|
Baertschiger RM, Serre-Beinier V, Morel P, Bosco D, Peyrou M, Clément S, Sgroi A, Kaelin A, Buhler LH, Gonelle-Gispert C. Fibrogenic potential of human multipotent mesenchymal stromal cells in injured liver. PLoS One 2009; 4:e6657. [PMID: 19684854 PMCID: PMC2722022 DOI: 10.1371/journal.pone.0006657] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Accepted: 07/07/2009] [Indexed: 12/26/2022] Open
Abstract
Multipotent mesenchymal stromal cells (MSC) are currently investigated clinically as cellular therapy for a variety of diseases. Differentiation of MSC toward endodermal lineages, including hepatocytes and their therapeutic effect on fibrosis has been described but remains controversial. Recent evidence attributed a fibrotic potential to MSC. As differentiation potential might be dependent of donor age, we studied MSC derived from adult and pediatric human bone marrow and their potential to differentiate into hepatocytes or myofibroblasts in vitro and in vivo. Following characterization, expanded adult and pediatric MSC were co-cultured with a human hepatoma cell line, Huh-7, in a hepatogenic differentiation medium containing Hepatocyte growth factor, Fibroblast growth factor 4 and oncostatin M. In vivo, MSC were transplanted into spleen or liver of NOD/SCID mice undergoing partial hepatectomy and retrorsine treatment. Expression of mesenchymal and hepatic markers was analyzed by RT-PCR, Western blot and immunohistochemistry. In vitro, adult and pediatric MSC expressed characteristic surface antigens of MSC. Expansion capacity of pediatric MSC was significantly higher when compared to adult MSC. In co-culture with Huh-7 cells in hepatogenic differentiation medium, albumin expression was more frequently detected in pediatric MSC (5/8 experiments) when compared to adult MSC (2/10 experiments). However, in such condition pediatric MSC expressed alpha smooth muscle more strongly than adult MSC. Stable engraftment in the liver was not achieved after intrasplenic injection of pediatric or adult MSC. After intrahepatic injection, MSC permanently remained in liver tissue, kept a mesenchymal morphology and expressed vimentin and alpha smooth muscle actin, but no hepatic markers. Further, MSC localization merges with collagen deposition in transplanted liver and no difference was observed using adult or pediatric MSC. In conclusion, when transplanted into an injured or regenerating liver, MSC differentiated into myofibroblasts with development of fibrous tissue, regardless of donor age. These results indicate that MSC in certain circumstances might be harmful due to their fibrogenic potential and this should be considered before potential use of MSC for cell therapy.
Collapse
Affiliation(s)
- Reto M. Baertschiger
- Surgical Research Unit, Department of Surgery, University Hospital Geneva, Geneva, Switzerland
| | - Véronique Serre-Beinier
- Surgical Research Unit, Department of Surgery, University Hospital Geneva, Geneva, Switzerland
| | - Philippe Morel
- Surgical Research Unit, Department of Surgery, University Hospital Geneva, Geneva, Switzerland
| | - Domenico Bosco
- Cell Isolation and Transplantation Center, Department of Surgery, University Hospital Geneva, Geneva, Switzerland
| | - Marion Peyrou
- Surgical Research Unit, Department of Surgery, University Hospital Geneva, Geneva, Switzerland
| | - Sophie Clément
- Department of Pathology and Immunology, Medical School of Geneva, Geneva, Switzerland
| | - Antonino Sgroi
- Surgical Research Unit, Department of Surgery, University Hospital Geneva, Geneva, Switzerland
| | - André Kaelin
- Department of Pediatric Orthopedics, Children's Hospital, University Hospital of Geneva, Geneva, Switzerland
| | - Leo H. Buhler
- Surgical Research Unit, Department of Surgery, University Hospital Geneva, Geneva, Switzerland
| | - Carmen Gonelle-Gispert
- Surgical Research Unit, Department of Surgery, University Hospital Geneva, Geneva, Switzerland
- * E-mail:
| |
Collapse
|
38
|
Wang H, Wang S, Hu J, Kong Y, Chen S, Li L, Li L. Oct4 is expressed in Nestin-positive cells as a marker for pancreatic endocrine progenitor. Histochem Cell Biol 2009; 131:553-63. [PMID: 19224238 DOI: 10.1007/s00418-009-0560-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2009] [Indexed: 01/18/2023]
Abstract
There are abundant progenitor cells in the developing pancreas, but molecular markers for these cells are lacking. Octamer-binding transcription factor-4 (Oct4) is an important transcription factor for keeping the features of self-renewal and pluripotency of embryonic stem cells. It's well known that Oct4, as a totipotent stem cells marker, just is expressed in totipotent stem cells. In the present study, we collected ten human fetal pancreases, and found that Oct4 mRNA and protein were expressed in human fetal pancreas samples by RT-PCR, western blot and immunohistochemistry assays. Using double-staining, we demonstrated that Oct4 was not co-expressed with Chromogranin A (a peptide expressed in endocrine cells), but partially co-expressed with Ngn3 (a transcription factor expressed in pancreatic endocrine precursor cells) and Nestin (a intermediate filament, Nestin-positive cells isolated from islets can be induced to express insulin) in human fetal pancreases. Indeed, we prepared Nestin-positive cells from human fetal pancreas by cell selection, and found that these cells expressed Oct4 and Ngn3. The Nestin-positive cells displayed a rapid duplication and could differentiate into osteoblasts, fat and endocrine cells in vitro. These results indicated that the Nestin-positive cells in the fetal age should be pancreatic progenitor cells. Overall, our study suggested that Oct4 was a marker for pancreatic endocrine progenitor.
Collapse
Affiliation(s)
- Hong Wang
- Stem Cell Research Center, Health Science Center, Peking University, No. 38 Xueyuan Road, Haidian District, Beijing, China.
| | | | | | | | | | | | | |
Collapse
|
39
|
Boumaza I, Srinivasan S, Witt WT, Feghali-Bostwick C, Dai Y, Garcia-Ocana A, Feili-Hariri M. Autologous bone marrow-derived rat mesenchymal stem cells promote PDX-1 and insulin expression in the islets, alter T cell cytokine pattern and preserve regulatory T cells in the periphery and induce sustained normoglycemia. J Autoimmun 2008; 32:33-42. [PMID: 19062254 DOI: 10.1016/j.jaut.2008.10.004] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Revised: 10/28/2008] [Accepted: 10/31/2008] [Indexed: 12/11/2022]
Abstract
Cell-based therapies offer considerable promise for prevention or cure of diabetes. We explored the potential of autologous, self-renewing, mesenchymal stem cells (MSC) as a clinically-applicable approach to promote glucose homeostasis. In vitro-expanded syngeneic bone marrow-derived MSC were administered following or prior to diabetes induction into a rat model of streptozotocin-induced beta cell injury. MSC were CD45(-)/CD44(+)/CD54(+)/CD90(+)/CD106(+). MSC spontaneously secreted IL-6, HGF, TGF-beta1 and expressed high levels of SDF-1 and low levels of VEGF, IL-1beta and PGE(2), but no EGF, insulin or glucagon. MSC homed to the pancreas and this therapy allowed for enhanced insulin secretion and sustained normoglycemia. Interestingly, immunohistochemistry demonstrated that, the islets from MSC-treated rats expressed high levels of PDX-1 and that these cells were also positive for insulin staining. In addition, peripheral T cells from MSC-treated rats exhibited a shift toward IL-10/IL-13 production and higher frequencies of CD4(+)/CD8(+) Foxp3(+) T cells compared to the PBS-treated rats. These data suggest that the bioactive factors secreted by MSC establish a tissue microenvironment that supports beta cell activation/survival in the pancreas. In addition, because of anti-inflammatory and immunoregulatory effects of MSC on T cells, this work can lead to clinical trial of autologous MSC to prevent/cure type-1 diabetes.
Collapse
Affiliation(s)
- Imene Boumaza
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | |
Collapse
|