1
|
Liu Y, Li P, Yang Y. Advancements in utilizing CD34 + stem cells for repairing diabetic vascular damage. Biochem Biophys Res Commun 2025; 750:151411. [PMID: 39889623 DOI: 10.1016/j.bbrc.2025.151411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Diabetes-related vascular damage is a frequent complication of diabetes that results in structural and functional impairment of blood vessels. This damage significantly heightens the risk of cardiovascular events. CD34+ stem cells have shown great potential in the treatment of diabetes-related vascular damage due to their differentiation and vascular repair capabilities. This article provides a review of the research hotspots on the role and mechanisms of CD34+ stem cells in the repair of diabetes-related vascular damage, including changes in cell quantity and function during diabetes, as well as the latest research on activating, protecting, or repairing these cells to prevent or treat vascular damage. The article also summarizes the impact of diabetes on the mobilization and function of CD34+ stem cells, emphasizing how diabetes negatively affects their ability to promote angiogenesis. These deficits can result in various complications, including issues with small blood vessels, coronary heart disease, foot problems, and retinal complications. On the clinical side, the article highlights the positive effects of CD34+ stem cell therapy in improving vascular function and tissue repair in diabetic patients, while also mentioning the inconsistencies in results between diabetes models and clinical studies, which necessitate further research to optimize treatment strategies. It emphasizes the importance of enhancing the mobilization, homing, and repair capabilities of CD34+ stem cells, as well as combining them with other treatment methods, to develop more effective strategies for treating diabetes-related vascular damage.
Collapse
Affiliation(s)
- Yiting Liu
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Pengyun Li
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.
| | - Yan Yang
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.
| |
Collapse
|
2
|
Mahajan N, Luo Q, Abhyankar S, Bhatwadekar AD. Transcriptomic Profile of Lin -Sca1 +c-kit (LSK) cells in db/db mice with long-standing diabetes. BMC Genomics 2024; 25:782. [PMID: 39134978 PMCID: PMC11318115 DOI: 10.1186/s12864-024-10679-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 08/01/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND The Lin-Sca1+c-Kit+ (LSK) fraction of the bone marrow (BM) comprises multipotent hematopoietic stem cells (HSCs), which are vital to tissue homeostasis and vascular repair. While diabetes affects HSC homeostasis overall, the molecular signature of mRNA and miRNA transcriptomic under the conditions of long-standing type 2 diabetes (T2D;>6 months) remains unexplored. METHODS In this study, we assessed the transcriptomic signature of HSCs in db/db mice, a well-known and widely used model for T2D. LSK cells of db/db mice enriched using a cell sorter were subjected to paired-end mRNA and single-end miRNA seq library and sequenced on Illumina NovaSeq 6000. The mRNA sequence reads were mapped using STAR (Spliced Transcripts Alignment to a Reference), and the miRNA sequence reads were mapped to the designated reference genome using the Qiagen GeneGlobe RNA-seq Analysis Portal with default parameters for miRNA. RESULTS We uncovered 2076 out of 13,708 mRNAs and 35 out of 191 miRNAs that were expressed significantly in db/db animals; strikingly, previously unreported miRNAs (miR-3968 and miR-1971) were found to be downregulated in db/db mice. Furthermore, we observed a molecular shift in the transcriptome of HSCs of diabetes with an increase in pro-inflammatory cytokines (Il4, Tlr4, and Tnf11α) and a decrease in anti-inflammatory cytokine IL10. Pathway mapping demonstrated inflammation mediated by chemokine, cytokine, and angiogenesis as one of the top pathways with a significantly higher number of transcripts in db/db mice. These molecular changes were reflected in an overt defect in LSK mobility in the bone marrow. miRNA downstream target analysis unveils several mRNAs targeting leukocyte migration, microglia activation, phagosome formation, and macrophage activation signaling as their primary pathways, suggesting a shift to an inflammatory phenotype. CONCLUSION Our findings highlight that chronic diabetes adversely alters HSCs' homeostasis at the transcriptional level, thus potentially contributing to the inflammatory phenotype of HSCs under long-term diabetes. We also believe that identifying HSCs-based biomarkers in miRNAs or mRNAs could serve as diagnostic markers and potential therapeutic targets for diabetes and associated vascular complications.
Collapse
Affiliation(s)
- Neha Mahajan
- Department of Ophthalmology, Indiana University School of Medicine, 1160 W Michigan St, Indianapolis, IN, 46202, USA
| | - Qianyi Luo
- Department of Ophthalmology, Indiana University School of Medicine, 1160 W Michigan St, Indianapolis, IN, 46202, USA
| | - Surabhi Abhyankar
- Department of Ophthalmology, Indiana University School of Medicine, 1160 W Michigan St, Indianapolis, IN, 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN, 46202, USA
| | - Ashay D Bhatwadekar
- Department of Ophthalmology, Indiana University School of Medicine, 1160 W Michigan St, Indianapolis, IN, 46202, USA.
| |
Collapse
|
3
|
Mahajan N, Luo Q, Abhyankar S, Bhatwadekar AD. Transcriptomic Profile of Lin - Sca1 + c-kit (LSK) cells in db/db mice with long-standing diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.576754. [PMID: 38328165 PMCID: PMC10849703 DOI: 10.1101/2024.01.22.576754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The Lin - Sca1 + c-Kit + (LSK) fraction comprises multipotent hematopoietic stem cells (HSCs), vital to tissue homeostasis and vascular repair. While HSC homeostasis is impaired in diabetes, it is not known how chronic (>6 months) type 2 diabetes (T2D) alters the HSC transcriptome. Herein, we assessed the transcriptomic signature of HSCs in db/db mice employing mRNA and miRNA sequencing. We uncovered 2076 mRNAs and 35 miRNAs differentially expressed in db/db mice, including two novel miRNAs previously unreported in T2D. Further analysis of these transcripts showed a molecular shift with an increase in the pro-inflammatory cytokines and a decrease in anti-inflammatory cytokine expression. Also, pathway mapping unveiled inflammation and angiogenesis as one of the top pathways. These effects were reflected in bone marrow mobilopathy, retinal microglial inflammation, and neurovascular deficits in db/db mice. In conclusion, our study highlights that chronic diabetes alters HSCs' at the transcriptomic level, thus potentially contributing to overall homeostasis and neurovascular deficits of diabetes, such as diabetic retinopathy. Highlights Bone marrow mobilopathy with long-standing diabetesSwitch in LSK transcriptomic profile towards inflammation and angiogenesisDiscovered 35 miRNAs, including two novel miRNAs, miR-3968 and miR-1971LSK dysfunction reflected in inflammation and neurovascular deficits of the retina.
Collapse
|
4
|
Çelik S, Kaynar L, Güven ZT, Begendi NK, Demir F, Keklik M, Ünal A. The impact of diabetes mellitus on hematopoietic stem cell mobilization, a-single center cohort study. Transfus Apher Sci 2023; 62:103838. [PMID: 37925340 DOI: 10.1016/j.transci.2023.103838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 11/06/2023]
Abstract
BACKGROUND Factors such as age, underlying hematological disease, chemotherapy and radiotherapy used, and bone marrow infiltration may cause mobilization failure. Several preclinical observed that diabetes mellitus (DM) leads to profound remodeling of the hematopoietic stem cell (HSC) niche, resulting in the impaired release of HSCs. We aim to examine the effect of DM on HSC mobilization and to investigate whether there is a relationship between complications developing in the DM process and drugs used to treat DM and mobilization failure. METHODS In Erciyes University Bone Marrow Transplantation Unit, 218 patients who underwent apheresis for stem cell mobilization between 2011 and 2021 were evaluated retrospectively. One hundred and nine patients had a diagnosis of DM, and 109 did not. RESULTS Mobilization failure developed in 17 (15.6 %) of the patients in the DM group, while it developed in 7 (6.4 %) patients in the non-DM group (p = 0.03). CD34+ stem cell count was 8.05 (1.3-30.2) × 106/kg in the DM group, while it was 8.2 (1.7-37.3) × 106/kg in the other group (p = 0.55). There was no statistically significant relationship between glucose and hemoglobin A1c levels and the amount of CD34+ cells (p = 0.83 and p = 0.14, respectively). Using sulfonylurea was the only independent predictor of mobilization failure (OR 5.75, 95 % CI: 1.38-24.05, p = 0.02). CONCLUSION DM should be considered a risk factor for mobilization failure. Further research is needed fully to understand the mechanisms underlying the mobilization failure effects of sulfonylureas and to develop strategies to improve stem cell mobilization in diabetic patients.
Collapse
Affiliation(s)
- Serhat Çelik
- Department of Hematology, Yenimahalle Training and Research Hospital, Yıldırım Beyazıt University, Ankara, Turkiye.
| | - Leylagül Kaynar
- Department of Hematology, Faculty of Medicine, Medipol Mega University, İstanbul, Turkiye
| | | | - Nermin Keni Begendi
- Department of Hematology, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkiye
| | - Fatma Demir
- Department of Medical Genetics, Ankara Bilkent City Hospital, Ankara, Turkiye
| | - Muzaffer Keklik
- Department of Hematology, Faculty of Medicine, Erciyes University, Kayseri, Turkiye
| | - Ali Ünal
- Department of Hematology, Faculty of Medicine, Erciyes University, Kayseri, Turkiye
| |
Collapse
|
5
|
Fadini GP, Albiero M. Impaired haematopoietic stem / progenitor cell traffic and multi-organ damage in diabetes. Stem Cells 2022; 40:716-723. [PMID: 35552468 PMCID: PMC9406601 DOI: 10.1093/stmcls/sxac035] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/04/2022] [Indexed: 11/18/2022]
Abstract
During antenatal development, hematopoietic stem/progenitor cells (HSPCs) arise from a specialized endothelium and migrate from the extraembryonic mesoderm to the fetal liver before establishing hematopoiesis in the bone marrow (BM). It is still debated whether, in adulthood, HSPCs display such ontologic overlap with vascular cells and capacity for endothelial differentiation. Yet, adult HSPCs retain a prominent migratory activity and traffic in the bloodstream to secondary lymphoid organs and all peripheral tissues, before eventually returning to the BM. While patrolling parenchymatous organs, HSPCs locate close to the vasculature, where they establish local hematopoietic islands and contribute to tissue homeostasis by paracrine signals. Solid evidence shows that diabetes mellitus jeopardizes the traffic of HSPCs from BM to the circulation and peripheral tissues, a condition called “mobilopathy.” A reduction in the levels of circulating HSPCs is the most immediate and apparent consequence, which has been consistently observed in human diabetes, and is strongly associated with future risk for multi-organ damage, including micro- and macro-angiopathy. But the shortage of HSPCs in the blood is only the visible tip of the iceberg. Abnormal HSPC traffic results from a complex interplay among metabolism, innate immunity, and hematopoiesis. Notably, mobilopathy is mechanistically connected with diabetes-induced myelopoiesis. Impaired traffic of HSPCs and enhanced generation of pro-inflammatory cells synergize for tissue damage and impair the resolution of inflammation. We herein summarize the current evidence that diabetes affects HSPC traffic, which are the causes and consequences of such alteration, and how it contributes to the overall disease burden.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Mattia Albiero
- Department of Medicine, University of Padova, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| |
Collapse
|
6
|
Perdomo S, Brugnini A, Trias N, Menyou A, Silveira G, Ranero S, Lens D, Díaz L, Grille S. Mobilized and apheresis-collected endothelial progenitor cells with plerixafor. J Clin Apher 2022; 37:245-252. [PMID: 35114004 DOI: 10.1002/jca.21967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) are immature cells able to proliferate and contribute to endothelial repair, vascular homeostasis, neovascularization, and angiogenesis. It therefore seems likely that circulating EPCs have therapeutic potential in ischemic and vascular diseases. In this study we evaluated the efficiency of EPC mobilization and collection by large volume leukapheresis in subjects with hematological diseases, treated with plerixafor in association with G-CSF. METHODS Twenty-two patients with lymphoid malignancies underwent rHuG-CSF and plerixafor treatment followed by leukapheresis. Blood samples before and after treatment and apheresis liquid sample were taken and analyzed by flow cytometry in order to quantified EPC. RESULTS The percentage of CD34+ cells and EPCs among circulating total nuclear cells (TNCs) increased significantly by approximately 2-fold and 3-fold, respectively, after plerixafor treatment. Consequently, the absolute number of CD34+ cells and EPCs were increased 4-fold after plerixafor treatment. The median PB concentration of EPCs before and after treatment were 0.77/μL (0.31-2.15) and 3.41/μL (1.78-4.54), respectively, P < .0001. The total EPCs collected per patient were 3.3×107 (0.8×107 -6.8×107 ). CONCLUSION We have shown that plerixafor in combination with G-CSF allows the mobilization and collection of large amounts of EPCs along with CD34+ cells in lymphoid neoplasm patients. The possibility to collect and to store these cells could represent a promising therapeutic tool for the treatment of ischemic complications without the need of in vitro expansion.
Collapse
Affiliation(s)
- Susana Perdomo
- Servicio Médico Integral, Centro de Trasplante de Médula Ósea, Montevideo, Uruguay
| | - Andreina Brugnini
- Laboratorio de Citometría y Biología Molecular, Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Natalia Trias
- Laboratorio de Citometría y Biología Molecular, Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Alba Menyou
- Servicio Médico Integral, Centro de Trasplante de Médula Ósea, Montevideo, Uruguay
| | - Gonzalo Silveira
- Laboratorio de Citometría y Biología Molecular, Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Sabrina Ranero
- Laboratorio de Citometría y Biología Molecular, Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.,Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Daniela Lens
- Laboratorio de Citometría y Biología Molecular, Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Lilián Díaz
- Servicio Médico Integral, Centro de Trasplante de Médula Ósea, Montevideo, Uruguay
| | - Sofía Grille
- Servicio Médico Integral, Centro de Trasplante de Médula Ósea, Montevideo, Uruguay.,Laboratorio de Citometría y Biología Molecular, Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.,Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
7
|
Salybekov AA, Wolfien M, Kobayashi S, Steinhoff G, Asahara T. Personalized Cell Therapy for Patients with Peripheral Arterial Diseases in the Context of Genetic Alterations: Artificial Intelligence-Based Responder and Non-Responder Prediction. Cells 2021; 10:3266. [PMID: 34943774 PMCID: PMC8699290 DOI: 10.3390/cells10123266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 01/14/2023] Open
Abstract
Stem/progenitor cell transplantation is a potential novel therapeutic strategy to induce angiogenesis in ischemic tissue, which can prevent major amputation in patients with advanced peripheral artery disease (PAD). Thus, clinicians can use cell therapies worldwide to treat PAD. However, some cell therapy studies did not report beneficial outcomes. Clinical researchers have suggested that classical risk factors and comorbidities may adversely affect the efficacy of cell therapy. Some studies have indicated that the response to stem cell therapy varies among patients, even in those harboring limited risk factors. This suggests the role of undetermined risk factors, including genetic alterations, somatic mutations, and clonal hematopoiesis. Personalized stem cell-based therapy can be developed by analyzing individual risk factors. These approaches must consider several clinical biomarkers and perform studies (such as genome-wide association studies (GWAS)) on disease-related genetic traits and integrate the findings with those of transcriptome-wide association studies (TWAS) and whole-genome sequencing in PAD. Additional unbiased analyses with state-of-the-art computational methods, such as machine learning-based patient stratification, are suited for predictions in clinical investigations. The integration of these complex approaches into a unified analysis procedure for the identification of responders and non-responders before stem cell therapy, which can decrease treatment expenditure, is a major challenge for increasing the efficacy of therapies.
Collapse
Affiliation(s)
- Amankeldi A. Salybekov
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, 1-1370 Okamoto, Kamakura 2478533, Japan;
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, 1-1370 Okamoto, Kamakura 2478533, Japan
| | - Markus Wolfien
- Department of Systems Biology and Bioinformatics, University of Rostock, Ulmenstrasse 69, 18057 Rostock, Germany;
| | - Shuzo Kobayashi
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, 1-1370 Okamoto, Kamakura 2478533, Japan;
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, 1-1370 Okamoto, Kamakura 2478533, Japan
| | - Gustav Steinhoff
- Department of Cardiac Surgery, Rostock University Medical Center, 18059 Rostock, Germany;
- Department Life, Light & Matter, University of Rostock, 18057 Rostock, Germany
| | - Takayuki Asahara
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, 1-1370 Okamoto, Kamakura 2478533, Japan
| |
Collapse
|
8
|
Taylor GS, Shaw A, Smith K, Capper TE, Scragg JH, Cronin M, Bashir A, Flatt A, Campbell MD, Stevenson EJ, Shaw JA, Ross M, West DJ. Type 1 diabetes patients increase CXCR4 + and CXCR7 + haematopoietic and endothelial progenitor cells with exercise, but the response is attenuated. Sci Rep 2021; 11:14502. [PMID: 34267242 PMCID: PMC8282661 DOI: 10.1038/s41598-021-93886-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/25/2021] [Indexed: 01/01/2023] Open
Abstract
Exercise mobilizes angiogenic cells, which stimulate vascular repair. However, limited research suggests exercise-induced increase of endothelial progenitor cell (EPCs) is completely lacking in type 1 diabetes (T1D). Clarification, along with investigating how T1D influences exercise-induced increases of other angiogenic cells (hematopoietic progenitor cells; HPCs) and cell surface expression of chemokine receptor 4 (CXCR4) and 7 (CXCR7), is needed. Thirty T1D patients and 30 matched non-diabetes controls completed 45 min of incline walking. Circulating HPCs (CD34+, CD34+CD45dim) and EPCs (CD34+VEGFR2+, CD34+CD45dimVEGFR2+), and subsequent expression of CXCR4 and CXCR7, were enumerated by flow cytometry at rest and post-exercise. Counts of HPCs, EPCs and expression of CXCR4 and CXCR7 were significantly lower at rest in the T1D group. In both groups, exercise increased circulating angiogenic cells. However, increases was largely attenuated in the T1D group, up to 55% lower, with CD34+ (331 ± 437 Δcells/mL vs. 734 ± 876 Δcells/mL p = 0.048), CD34+VEGFR2+ (171 ± 342 Δcells/mL vs. 303 ± 267 Δcells/mL, p = 0.006) and CD34+VEGFR2+CXCR4+ (126 ± 242 Δcells/mL vs. 218 ± 217 Δcells/mL, p = 0.040) significantly lower. Exercise-induced increases of angiogenic cells is possible in T1D patients, albeit attenuated compared to controls. Decreased mobilization likely results in reduced migration to, and repair of, vascular damage, potentially limiting the cardiovascular benefits of exercise.Trial registration: ISRCTN63739203.
Collapse
Affiliation(s)
- Guy S Taylor
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Andy Shaw
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Kieran Smith
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Tess E Capper
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK.,Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Jadine H Scragg
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK.,Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Michael Cronin
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Ayat Bashir
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Anneliese Flatt
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Matthew D Campbell
- Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland, UK.,Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Emma J Stevenson
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - James A Shaw
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Mark Ross
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, UK
| | - Daniel J West
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
9
|
So HS, Kim MG, Lee JC, Kook SH. Glucose oxidase induces mobilization of long-term repopulating hematopoietic cells in mice. Stem Cells Transl Med 2021; 10:1446-1453. [PMID: 34160898 PMCID: PMC8459634 DOI: 10.1002/sctm.20-0514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 05/16/2021] [Accepted: 06/06/2021] [Indexed: 12/17/2022] Open
Abstract
Hematopoietic stem progenitor cells (HSPCs) mobilized to peripheral blood, rather than those remaining in the bone marrow (BM), are commonly used as stem cell source in the clinic. As reactive oxygen species (ROS) are suggested as mediator of HSPC mobilization, we examined the impacts of glucose oxidase (GO) on peripheral mobilization of BM HSPCs and the associated mechanisms. Intravenous injection of GO induced HSPC mobilization even by single treatment, and the GO‐mobilized cells maintained their long‐term reconstituting and differentiating potentials in conditioned recipients. GO‐injected mice lived a normal life without adverse effects such as stem cell senescence, hematopoietic disorders, and blood parameter alteration. The mobilization effect of GO was even evident in animal models showing poor mobilization, such as old, 5‐fluorouracil‐treated, or alendronate‐treated mice. Importantly, combined injection of GO with granulocyte colony‐stimulating factor (G‐CSF) and/or AMD3100 enhanced more greatly HSPC mobilization than did G‐CSF, AMD3100, or both. The GO‐stimulated HSPC mobilization was almost completely attenuated by n‐acetyl‐L‐cysteine treatment. Collectively, our results not only highlight the potential role of GO in HSPC mobilization via ROS signaling, but also provide a GO‐based new strategy to improve HSPC mobilization in poorly mobilizing allogeneic or autologous donors via combination with G‐CSF and/or AMD3100.
Collapse
Affiliation(s)
- Han-Sol So
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea
| | - Min-Guk Kim
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea.,Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, South Korea
| | - Jeong-Chae Lee
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea.,Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, South Korea
| | - Sung-Ho Kook
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea
| |
Collapse
|
10
|
Lin X, Wang H, Li X. Mobilization of endothelial progenitor cells promotes angiogenesis after full thickness excision by AMD3100 combined with G-CSF in diabetic mice by SDF-1/CXCR4 axis. Diab Vasc Dis Res 2021; 18:14791641211002473. [PMID: 33779350 PMCID: PMC8481732 DOI: 10.1177/14791641211002473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
AIM The aim of the present study was to investigate the effect of the mobilization of EPCs by AMD3100 combined with G-CSF on wound healing in diabetic mice. METHODS The full-thickness excisional wounds model of diabetic mice (db/db) was examined by hematoxylin and eosin staining, immunohistochemical staining, and western blotting to compare the wound healing and neovascularization among the combination, AMD3100 alone, G-CSF alone, and control groups. RESULTS The wounds reached the complete closure in the combination, AMD3100 alone, G-CSF alone, and control groups on days 17, 20, 21, 21 after surgery, respectively. In addition, the combination group promoted the inflammatory cell recruitment and glandular formation. On day 10 from injury, the protein expression of CD31 in the combination group was significantly higher compared with the other three groups (p < 0.001). The level of SDF-1 protein remained high in the combined group until on day 10 after surgery (p < 0.001). CONCLUSION The mobilization of endogenous EPCs by AMD3100 combine with G-CSF is able to enhance the complete healing of full-thickness wounds and neovascularization in db/db mice may by SDF-1/CXCR4 axis. These findings provided a novel method and indication of duration of mobilization on diabetic wound healing and tissue regeneration.
Collapse
Affiliation(s)
- Xiaoying Lin
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hong Wang
- Department of Burns, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiaolan Li
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
11
|
Kh S, Haider KH. Stem Cells: A Renewable Source of Pancreatic β-Cells and Future for Diabetes Treatment. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
12
|
Albiero M, Bonora BM, Fadini GP. Diabetes pharmacotherapy and circulating stem/progenitor cells. State of the art and evidence gaps. Curr Opin Pharmacol 2020; 55:151-156. [PMID: 33271409 DOI: 10.1016/j.coph.2020.10.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022]
Abstract
Diabetes is burdened with the development of several end-organ complications leading to excess mortality. Though the causes of such organ damage are far from being clarified, diabetes has been redefined as a disease of impaired damage control, wherein ongoing damage is not adequately compensated by activation of repair processes. Bone marrow-derived hematopoietic stem/progenitor cells (HSPCs) and their descendants endothelial progenitor cells (EPCs) have been extensively studied as major players in tissue homeostasis as well as biomarkers of diabetic complication risk. Thus, strategies to raise the levels of circulating HSPCs/EPCs have attracted interest for their potential to modify the future risk of complications. We herein discuss state-of-the-art of the effects exerted by diabetes pharmacotherapy on such cell populations. Further, we highlight which outstanding questions remain to be addressed for a more comprehensive understanding of this topic.
Collapse
Affiliation(s)
- Mattia Albiero
- Department of Medicine, University of Padova, 35128 Padova, Italy; Veneto Institute of Molecular Medicine, 35128 Padova, Italy
| | - Benedetta Maria Bonora
- Department of Medicine, University of Padova, 35128 Padova, Italy; Veneto Institute of Molecular Medicine, 35128 Padova, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, 35128 Padova, Italy; Veneto Institute of Molecular Medicine, 35128 Padova, Italy.
| |
Collapse
|
13
|
Fadini GP, Mehta A, Dhindsa DS, Bonora BM, Sreejit G, Nagareddy P, Quyyumi AA. Circulating stem cells and cardiovascular outcomes: from basic science to the clinic. Eur Heart J 2020; 41:4271-4282. [PMID: 31891403 PMCID: PMC7825095 DOI: 10.1093/eurheartj/ehz923] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 10/19/2019] [Accepted: 12/05/2019] [Indexed: 02/06/2023] Open
Abstract
The cardiovascular and haematopoietic systems have fundamental inter-relationships during development, as well as in health and disease of the adult organism. Although haematopoietic stem cells (HSCs) emerge from a specialized haemogenic endothelium in the embryo, persistence of haemangioblasts in adulthood is debated. Rather, the vast majority of circulating stem cells (CSCs) is composed of bone marrow-derived HSCs and the downstream haematopoietic stem/progenitors (HSPCs). A fraction of these cells, known as endothelial progenitor cells (EPCs), has endothelial specification and vascular tropism. In general, the levels of HSCs, HSPCs, and EPCs are considered indicative of the endogenous regenerative capacity of the organism as a whole and, particularly, of the cardiovascular system. In the last two decades, the research on CSCs has focused on their physiologic role in tissue/organ homoeostasis, their potential application in cell therapies, and their use as clinical biomarkers. In this review, we provide background information on the biology of CSCs and discuss in detail the clinical implications of changing CSC levels in patients with cardiovascular risk factors or established cardiovascular disease. Of particular interest is the mounting evidence available in the literature on the close relationships between reduced levels of CSCs and adverse cardiovascular outcomes in different cohorts of patients. We also discuss potential mechanisms that explain this association. Beyond CSCs' ability to participate in cardiovascular repair, levels of CSCs need to be interpreted in the context of the broader connections between haematopoiesis and cardiovascular function, including the role of clonal haematopoiesis and inflammatory myelopoiesis.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Anurag Mehta
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, USA
| | - Devinder Singh Dhindsa
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, USA
| | | | - Gopalkrishna Sreejit
- Division of Cardiac Surgery, Department of Surgery, Ohio State University, Columbus, OH 43210, USA
| | - Prabhakara Nagareddy
- Division of Cardiac Surgery, Department of Surgery, Ohio State University, Columbus, OH 43210, USA
| | - Arshed Ali Quyyumi
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, USA
| |
Collapse
|
14
|
Chen J, Lazarus HM, Dahi PB, Avecilla S, Giralt SA. Getting blood out of a stone: Identification and management of patients with poor hematopoietic cell mobilization. Blood Rev 2020; 47:100771. [PMID: 33213986 DOI: 10.1016/j.blre.2020.100771] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 07/15/2020] [Accepted: 10/14/2020] [Indexed: 12/13/2022]
Abstract
Hematopoietic cell transplantation (HCT) has become a primary treatment for many cancers. Nowadays, the primary source of hematopoietic cells is by leukapheresis collection of these cells from peripheral blood, after a forced egress of hematopoietic cells from marrow into blood circulation, a process known as "mobilization". In this process, mobilizing agents disrupt binding interactions between hematopoietic cells and marrow microenvironment to facilitate collection. As the first essential step of HCT, poor mobilization, i.e. failure to obtain a desired or required number of hematopoietic cell, is one of the major factors affecting engraftment or even precluding transplantation. This review summarizes the available mobilization regimens using granulocyte-colony stimulating factor (G-CSF) and plerixafor, as well as the current understanding of the factors that are associated with poor mobilization. Strategies to mobilize patients or healthy donors who failed previous mobilization are discussed. Multiple novel agents are under investigation and some of them have shown the potential to enhance the mobilization response to G-CSF and/or plerixafor. Further investigation of the risk factors including genetic factors will offer an opportunity to better understand the molecular mechanism of mobilization and help develop new therapeutic strategies for successful mobilizations.
Collapse
Affiliation(s)
- Jian Chen
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States; Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Hillard M Lazarus
- Department of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Parastoo B Dahi
- Department of Medicine, Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Scott Avecilla
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Sergio A Giralt
- Department of Medicine, Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States.
| |
Collapse
|
15
|
Cappellari R, D'Anna M, Menegazzo L, Bonora BM, Albiero M, Avogaro A, Fadini GP. Diabetes mellitus impairs circulating proangiogenic granulocytes. Diabetologia 2020; 63:1872-1884. [PMID: 32306097 DOI: 10.1007/s00125-020-05142-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/28/2020] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS Cardiovascular risk in diabetes is at least in part attributable to defective angiogenesis. Since diabetes negatively affects blood cells involved in angiogenesis, we herein evaluated whether diabetes impairs proangiogenic granulocytes (PAGs). METHODS We characterised and quantified PAGs as CD49d+ granulocytes in peripheral blood of participants with type 2 or type 1 diabetes and in non-diabetic control participants. We evaluated PAG antigenic profile and assessed in vitro functional properties of CD49d+ granulocytes using 2D and 3D angiogenesis assays. We also quantified PAGs before and after glucose control with a sodium-glucose cotransporter 2 (SGLT2) inhibitor, dapagliflozin. In parallel, we measured Ly6G+CD49d+ PAGs in streptozotocin-induced type 1-like diabetic mice vs non-diabetic control mice. RESULTS PAGs were composed of eosinophils (>80%) and neutrophils (<20%). Within both populations, CD49d identified CXCR4high/VEGFR1high cells. CD49d+ granulocytes supported in vitro angiogenesis by endothelial cells significantly more than CD49d- control granulocytes, and physically interacted with endothelial cells. Granulocytes from type 2 diabetic participants had a profoundly impaired capacity to stimulate endothelial cell tubule formation compared with those from non-diabetic control participants. CD49d+ PAGs were reduced by 30-40% and were functionally impaired in diabetic vs control individuals. PAG levels inversely correlated with plasma glucose (r = -0.25; p = 0.025) and significantly increased 1.8-times after glucose control with dapagliflozin, which reduced HbA1c by 1.0% (11 mmol/mol). Levels of Ly6G+CD49d+ PAGs were also significantly reduced also in type 1 diabetic mice vs control mice. CONCLUSIONS/INTERPRETATION We illustrate a significant impairment of PAGs in diabetes and provide evidence for a direct role of hyperglycaemia. These findings add mechanistic information to explain the defective angiogenesis in diabetes. Graphical abstract.
Collapse
Affiliation(s)
- Roberta Cappellari
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128, Padova, Italy
| | - Marianna D'Anna
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128, Padova, Italy
| | - Lisa Menegazzo
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128, Padova, Italy
| | - Benedetta Maria Bonora
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128, Padova, Italy
| | - Mattia Albiero
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128, Padova, Italy
| | - Angelo Avogaro
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128, Padova, Italy
| | - Gian Paolo Fadini
- Veneto Institute of Molecular Medicine, Padova, Italy.
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128, Padova, Italy.
| |
Collapse
|
16
|
Wei W, Li L, Deng L, Wang ZJ, Dong JJ, Lyu XY, Jia T, Wang L, Wang HX, Mao H, Zhao S. Autologous Bone Marrow Mononuclear Cell Transplantation Therapy Improved Symptoms in Patients with Refractory Diabetic Sensorimotor Polyneuropathy via the Mechanisms of Paracrine and Immunomodulation: A Controlled Study. Cell Transplant 2020; 29:963689720949258. [PMID: 32787571 PMCID: PMC7563922 DOI: 10.1177/0963689720949258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
We recently reported that transplantation of autologous bone marrow mononuclear
cells (BM-MNCs) may be an effective and promising therapy to treat refractory
diabetic sensorimotor polyneuropathy (DSPN) in patients with type 2 diabetes
mellitus (T2DM). This study was designed to investigate the potential mechanisms
of BM-MNCs therapy, which recruited 60 patients with DSPN, 30 T2DM patients
without complications, and 30 healthy control participants. All clinical
parameters, the levels of inflammatory markers, and growth factors in the three
groups were compared. Patients in DSPN group had higher level of tumor necrosis
factor-α (TNF-α) (DSPN vs control, 412.90 ± 64.58 vs 374.81 ± 63.18 pg/mL,
P < 0.01) and lower level of vascular endothelial growth
factor (VEGF) (DSPN vs control, 140.93 ± 24.78 vs 157.39 ± 25.11 pg/mL,
P < 0.01) than those in control group. DSPN group had
the highest level of soluble intercellular adhesion molecule-1 (sICAM-1) among
three groups (DSPN and DM vs control, 1477.56 ± 228.00 and 1342.17 ± 237.54 vs
1308.00 ± 200.94 ng/mL, P < 0.05). The level of nerve growth
factor in the DSPN group was slightly lower than that in the DM group (DSPN vs
DM, 3509.11 ± 438.39 vs 3734.87 ± 647.50 pg/mL, P < 0.05).
All patients with DSPN received one intramuscular injection of BM-MNCs and
clinical follow-ups after the therapy for 2 days, 1, 4, 12, 24, and 48 weeks.
Neuropathic symptoms of foot pain, numbness, and weakness were significantly
improved within 4 weeks after BM-MNCs injection. Patients with DSPN were divided
into the responder (n = 35) and nonresponder groups
(n = 19) based on the improvement of nerve conduction
velocity at 12 weeks post-transplantation. Compared with nonresponders,
responders were younger (57.3 ± 5.2 vs 62.0 ± 4.8, P <
0.01), had a shorter history of diabetes (7.1 ± 2.7 vs 11.2 ± 5.4 years,
P < 0.01), and had higher numbers of mobilized
CD34+ cells (17.61 ± 2.64 vs 14.79 ± 1.62 ×105/L,
P < 0.01) and BM-MNCs (12.05 ± 2.16 vs 9.84 ± 1.53
×108/L, P < 0.01). The levels of TNF-α and
sICAM-1 decreased just after BM-MNCs injection in both groups and slowly
reverted to baseline levels. The duration of the downtrend of TNF-α and sICAM-1
in the responder group lasted longer than that in the nonresponder group. Serum
level of VEGF in the responder group increased immediately after BM-MNC therapy
and reached the highest point after the injection for 12 weeks. On the other
hand, VEGF levels in the nonresponder group only increased slightly. Binary
logistic regression was performed to evaluate the corresponding prognostic
factors for BM-MNCs treatment. The number of applied CD34+ cells and
the duration of diabetes were the independent predictors of responding to
BM-MNCs therapy. No adverse event associated with the treatment was observed
during follow-up observations. These results indicated that BM-MNCs
transplantation is an effective and promising therapeutic strategy to treat
refractory DSPN. The immune regulation and paracrine function of BM-MNCs may
contribute to the improvement of DSPN.
Collapse
Affiliation(s)
- Wei Wei
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, 12403Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Li Li
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, 12403Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lin Deng
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, 12403Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhong-Jing Wang
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, 12403Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jing-Jian Dong
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, 12403Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiao-Yu Lyu
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, 12403Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ting Jia
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, 12403Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Li Wang
- Department of Hematology, The Central Hospital of Wuhan, Tongji Medical College, 12403Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hong-Xiang Wang
- Department of Hematology, The Central Hospital of Wuhan, Tongji Medical College, 12403Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hong Mao
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, 12403Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shi Zhao
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, 12403Huazhong University of Science and Technology, Wuhan, Hubei, China.,Regenerative Medical Center of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, 12403Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
17
|
Tedesco S, Ciciliot S, Menegazzo L, D'Anna M, Scattolini V, Cappellari R, Cignarella A, Avogaro A, Albiero M, Fadini GP. Pharmacologic PPAR-γ Activation Reprograms Bone Marrow Macrophages and Partially Rescues HSPC Mobilization in Human and Murine Diabetes. Diabetes 2020; 69:1562-1572. [PMID: 32345753 DOI: 10.2337/db19-0640] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 04/22/2020] [Indexed: 11/13/2022]
Abstract
Mobilization of hematopoietic stem/progenitor cells (HSPC) from the bone marrow (BM) is impaired in diabetes. Excess oncostatin M (OSM) produced by M1 macrophages in the diabetic BM signals through p66Shc to induce Cxcl12 in stromal cells and retain HSPC. BM adipocytes are another source of CXCL12 that blunts mobilization. We tested a strategy of pharmacologic macrophage reprogramming to rescue HSPC mobilization. In vitro, PPAR-γ activation with pioglitazone switched macrophages from M1 to M2, reduced Osm expression, and prevented transcellular induction of Cxcl12 In diabetic mice, pioglitazone treatment downregulated Osm, p66Shc, and Cxcl12 in the hematopoietic BM, restored the effects of granulocyte-colony stimulation factor (G-CSF), and partially rescued HSPC mobilization, but it increased BM adipocytes. Osm deletion recapitulated the effects of pioglitazone on adipogenesis, which was p66Shc independent, and double knockout of Osm and p66Shc completely rescued HSPC mobilization. In the absence of OSM, BM adipocytes produced less CXCL12, being arguably devoid of HSPC-retaining activity, whereas pioglitazone failed to downregulate Cxcl12 in BM adipocytes. In patients with diabetes on pioglitazone therapy, HSPC mobilization after G-CSF was partially rescued. In summary, pioglitazone reprogrammed BM macrophages and suppressed OSM signaling, but sustained Cxcl12 expression by BM adipocytes could limit full recovery of HSPC mobilization.
Collapse
Affiliation(s)
- Serena Tedesco
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
| | | | | | - Marianna D'Anna
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
| | - Valentina Scattolini
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
| | - Roberta Cappellari
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
| | | | - Angelo Avogaro
- Department of Medicine, University of Padova, Padova, Italy
| | - Mattia Albiero
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
| | - Gian Paolo Fadini
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
| |
Collapse
|
18
|
Bonora BM, Cappellari R, Mazzucato M, Rigato M, Grasso M, Menegolo M, Bruttocao A, Avogaro A, Fadini GP. Stem cell mobilization with plerixafor and healing of diabetic ischemic wounds: A phase IIa, randomized, double-blind, placebo-controlled trial. Stem Cells Transl Med 2020; 9:965-973. [PMID: 32485785 PMCID: PMC7445026 DOI: 10.1002/sctm.20-0020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/25/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
Bone marrow‐derived cells contribute to tissue repair, but traffic of hematopoietic stem/progenitor cells (HSPCs) is impaired in diabetes. We therefore tested whether HSPC mobilization with the CXCR4 antagonist plerixafor improved healing of ischemic diabetic wounds. This was a pilot, phase IIa, double‐blind, randomized, placebo‐controlled trial (NCT02790957). Patients with diabetes with ischemic wounds were randomized to receive a single subcutaneous injection of plerixafor or saline on top of standard medical and surgical therapy. The primary endpoint was complete healing at 6 months. Secondary endpoints were wound size, transcutaneous oxygen tension (TcO2), ankle‐brachial index (ABI), amputations, and HSPC mobilization. Twenty‐six patients were enrolled: 13 received plerixafor and 13 received placebo. Patients were 84.6% males, with a mean age of 69 years. HSPC mobilization was successful in all patients who received plerixafor. The trial was terminated after a preplanned interim analysis of 50% of the target population showed a significantly lower healing rate in the plerixafor vs the placebo group. In the final analysis data set, the rate of complete healing was 38.5% in the plerixafor group vs 69.2% in the placebo group (chi‐square P = .115). Wound size tended to be larger in the plerixafor group for the entire duration of observation. No significant difference was noted for the change in TcO2 and ABI or in amputation rates. No other safety concern emerged. In conclusion, successful HSPC mobilization with plerixafor did not improve healing of ischemic diabetic wounds. Contrary to what was expected, outside the context of hematological disorders, mobilization of diabetic HSPCs might exert adverse effects on wound healing.
Collapse
Affiliation(s)
- Benedetta Maria Bonora
- Department of Medicine, University of Padova, Padova, Italy.,Veneto Institute of Molecular Medicine, Padova, Italy
| | - Roberta Cappellari
- Department of Medicine, University of Padova, Padova, Italy.,Veneto Institute of Molecular Medicine, Padova, Italy
| | | | - Mauro Rigato
- Department of Medicine, University of Padova, Padova, Italy.,ULSS2 Diabetology Service, Treviso, Italy
| | - Marco Grasso
- Department of Medicine, University of Padova, Padova, Italy
| | - Mirko Menegolo
- Department of Cardiothoracic and Vascular Sciences, University of Padova, Padova, Italy
| | | | - Angelo Avogaro
- Department of Medicine, University of Padova, Padova, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Padova, Italy.,Veneto Institute of Molecular Medicine, Padova, Italy.,ULSS2 Diabetology Service, Treviso, Italy
| |
Collapse
|
19
|
Sun L, Xi S, He G, Li Z, Gang X, Sun C, Guo W, Wang G. Two to Tango: Dialogue between Adaptive and Innate Immunity in Type 1 Diabetes. J Diabetes Res 2020; 2020:4106518. [PMID: 32802890 PMCID: PMC7415089 DOI: 10.1155/2020/4106518] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/18/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a long-term and chronic autoimmune disorder, in which the immune system attacks the pancreatic β-cells. Both adaptive and innate immune systems are involved in T1DM development. Both B-cells and T-cells, including CD4 + and CD8 + T-cells, as well as other T-cell subsets, could affect onset of autoimmunity. Furthermore, cells involved in innate immunity, including the macrophages, dendritic cells, and natural killer (NK) cells, could also accelerate or decelerate T1DM development. In this review, the crosstalk and function of immune cells in the pathogenesis of T1DM, as well as the corresponding therapeutic interventions, are discussed.
Collapse
Affiliation(s)
- Lin Sun
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021 Jilin, China
| | - Shugang Xi
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021 Jilin, China
| | - Guangyu He
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021 Jilin, China
| | - Zhuo Li
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021 Jilin, China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021 Jilin, China
| | - Chenglin Sun
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021 Jilin, China
| | - Weiying Guo
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021 Jilin, China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021 Jilin, China
| |
Collapse
|
20
|
Golay H, Jurkovic Mlakar S, Mlakar V, Nava T, Ansari M. The Biological and Clinical Relevance of G Protein-Coupled Receptors to the Outcomes of Hematopoietic Stem Cell Transplantation: A Systematized Review. Int J Mol Sci 2019; 20:E3889. [PMID: 31404983 PMCID: PMC6719093 DOI: 10.3390/ijms20163889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/06/2019] [Accepted: 08/07/2019] [Indexed: 01/04/2023] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) remains the only curative treatment for several malignant and non-malignant diseases at the cost of serious treatment-related toxicities (TRTs). Recent research on extending the benefits of HSCT to more patients and indications has focused on limiting TRTs and improving immunological effects following proper mobilization and engraftment. Increasing numbers of studies report associations between HSCT outcomes and the expression or the manipulation of G protein-coupled receptors (GPCRs). This large family of cell surface receptors is involved in various human diseases. With ever-better knowledge of their crystal structures and signaling dynamics, GPCRs are already the targets for one third of the current therapeutic arsenal. The present paper assesses the current status of animal and human research on GPCRs in the context of selected HSCT outcomes via a systematized survey and analysis of the literature.
Collapse
Affiliation(s)
- Hadrien Golay
- Platform of Pediatric Onco-Hematology research (CANSEARCH Laboratory), Department of Pediatrics, Gynecology, and Obstetrics, University of Geneva, Bâtiment La Tulipe, Avenue de la Roseraie 64, 1205 Geneva, Switzerland
| | - Simona Jurkovic Mlakar
- Platform of Pediatric Onco-Hematology research (CANSEARCH Laboratory), Department of Pediatrics, Gynecology, and Obstetrics, University of Geneva, Bâtiment La Tulipe, Avenue de la Roseraie 64, 1205 Geneva, Switzerland
| | - Vid Mlakar
- Platform of Pediatric Onco-Hematology research (CANSEARCH Laboratory), Department of Pediatrics, Gynecology, and Obstetrics, University of Geneva, Bâtiment La Tulipe, Avenue de la Roseraie 64, 1205 Geneva, Switzerland
| | - Tiago Nava
- Platform of Pediatric Onco-Hematology research (CANSEARCH Laboratory), Department of Pediatrics, Gynecology, and Obstetrics, University of Geneva, Bâtiment La Tulipe, Avenue de la Roseraie 64, 1205 Geneva, Switzerland
- Department of Women-Children-Adolescents, Division of General Pediatrics, Pediatric Onco-Hematology Unit, Geneva University Hospitals (HUG), Avenue de la Roseraie 64, 1205 Geneva, Switzerland
| | - Marc Ansari
- Platform of Pediatric Onco-Hematology research (CANSEARCH Laboratory), Department of Pediatrics, Gynecology, and Obstetrics, University of Geneva, Bâtiment La Tulipe, Avenue de la Roseraie 64, 1205 Geneva, Switzerland.
- Department of Women-Children-Adolescents, Division of General Pediatrics, Pediatric Onco-Hematology Unit, Geneva University Hospitals (HUG), Avenue de la Roseraie 64, 1205 Geneva, Switzerland.
| |
Collapse
|
21
|
Mao H, Wei W, Fu XL, Dong JJ, Lyu XY, Jia T, Tang Y, Zhao S. Efficacy of autologous bone marrow mononuclear cell transplantation therapy in patients with refractory diabetic peripheral neuropathy. Chin Med J (Engl) 2019; 132:11-16. [PMID: 30628954 PMCID: PMC6629317 DOI: 10.1097/cm9.0000000000000009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Owing to the multifactorial nature of the pathogenesis of diabetic peripheral neuropathy (DPN), conventional drug therapies have not been effective. The application of stem cells transplantation may be useful for the treatment of DPN. This study was designed to assess the safety and therapeutic effects of autologous bone marrow mononuclear cells (BMMNCs) transplantation on the treatment of refractory DPN. METHODS One hundred and sixty-eight patients with refractory DPN were recruited and enrolled in the study. They received intramuscular injection of BMMNCs and followed at 1, 3, 6, 12, 18, 24, and 36 months after the transplantation. Clinical data, Toronto Clinical Scoring System (TCSS), and nerve conduction studies (NCSs) were compared before and after the transplantation. RESULTS The signs and symptoms of neuropathy were significantly improved after BMMNCs transplantation. The values of the TCSS scores at 1 month (9.68 ± 2.49 vs. 12.55 ± 2.19, P < 0.001) and 3 months (8.47 ± 2.39 vs. 12.55 ± 2.19, P < 0.001) after the treatment reduced significantly compared with the baseline value. This decrement remained persistent until the end of the study. The conduction velocity and action potential and sensory nerves were significantly improved after transplantation (3 and 12 months after the treatment vs. the baseline: motor nerve conduction velocity, 40.24 ± 2.80 and 41.00 ± 2.22 m/s vs. 38.21 ± 2.28 m/s, P < 0.001; sensory nerve conduction velocity, 36.96 ± 2.26 and 39.15 ± 2.61 m/s vs. 40.41 ± 2.22 m/s, P < 0.001; compound muscle action potential, 4.67 ± 1.05 and 5.50 ± 1.20 μV vs. 5.68 ± 1.08 μV, P < 0.001; sensory nerve action potential, 4.29 ± 0.99 and 5.14 ± 1.26 μV vs. 5.41 ± 1.14 μV, P < 0.001). No adverse event associated with the treatment was observed during the follow-up period. CONCLUSIONS Autologous transplantation of BMMNCs may be an effective and promising therapeutic strategy for the treatment of refractory DPN.
Collapse
Affiliation(s)
- Hong Mao
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Wei Wei
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Xiu-Li Fu
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Jing-Jian Dong
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Xiao-Yu Lyu
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Ting Jia
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Yang Tang
- Department of Economics, Nanyang Technological University, Singapore 637332, Singapore
| | - Shi Zhao
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| |
Collapse
|
22
|
Kim JY, Lee JY, Ha KS, Han ET, Park WS, Min CK, Hong SH. Perivascular Cells and NADPH Oxidase Inhibition Partially Restore Hyperglycemia-Induced Alterations in Hematopoietic Stem Cell and Myeloid-Derived Suppressor Cell Populations in the Bone Marrow. Int J Stem Cells 2019; 12:63-72. [PMID: 30595009 PMCID: PMC6457702 DOI: 10.15283/ijsc18097] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 10/19/2018] [Accepted: 11/25/2018] [Indexed: 12/31/2022] Open
Abstract
Background and Objectives Patients suffer from long-term diabetes can result in severe complications in multiple organs through induction of vascular dysfunctions. However, the effects of chronic hyperglycemic conditions on hematopoiesis and the microenvironment in the bone marrow (BM) are not yet well understood. Methods BM cells were harvested from femurs of mice and analyzed using flow cytometry. Human PVCs were cultured in serum-free α-MEM. After 24hrs, PVC-CM was collected and filtered through a 0.22 μm filter. Results In this study, we showed that hyperglycemia alters hematopoietic composition in the BM, which can partially be restored via paracrine mechanisms, including perivascular cells (PVCs) and NADPH oxidase (NOX) inhibition in mice with streptozotocin-induced diabetes. Prolonged hyperglycemic conditions resulted in an increase in the frequency and number of long-term hematopoietic stem cells as well as the number of total BM cells. The altered hematopoiesis in the BM was partially recovered by treatment with PVC-derived conditioned medium (CM). Long-term diabetes also increased the number of myeloid-derived suppressor cells in the BM, which was partially restored by the administration of PVC-CM and diphenyleneiodonium (DPI), a NOX inhibitor. We further showed the downregulation of ERK and p38 phosphorylation in BM cells of diabetic mice treated with PVC-CM and DPI. This may be associated with dysfunction of hematopoietic cells and promotion of subsequent diabetic complications. Conclusions Our data suggested that alterations in BM hematopoietic composition due to prolonged hyperglycemic conditions might be restored by improvement of the hematopoietic microenvironment and modulation of NOX activity.
Collapse
Affiliation(s)
- Ji-Young Kim
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Ji Yoon Lee
- Department of Biomedical Sciences, Stem Cell Institute, CHA University, Seongnam, Korea
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Won Sun Park
- Department of Physiology, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Chang-Ki Min
- Department of Hematology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.,Leukemia Research Institute, The Catholic University of Korea, Seoul, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Korea
| |
Collapse
|
23
|
Khan J, Alexander A, Agrawal M, Ajazuddin, Dubey SK, Siddique S, Saraf S, Saraf S. Stem Cell-Based Therapies: A New Ray of Hope for Diabetic Patients. Curr Stem Cell Res Ther 2019; 14:146-151. [DOI: 10.2174/1574888x13666181002154110] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 05/09/2018] [Accepted: 07/07/2018] [Indexed: 11/22/2022]
Abstract
Diabetes and its complications are a significant health concern throughout the globe. There are physiological differences in the mechanism of type-I and type-II diabetes and the conventional drug therapy as well as insulin administration seem to be insufficient to address the problem at large successfully. Hypoglycemic swings, frequent dose adjustments and resistance to the drug are major problems associated with drug therapy. Cellular approaches through stem cell based therapeutic interventions offer a promising solution to the problem. The need for pancreatic transplants in case of Type- I diabetes can also be by-passed/reduced due to the formation of insulin producing β cells via stem cells. Embryonic Stem Cells (ESCs) and induced Pluripotent Stem Cells (iPSCs), successfully used for generating insulin producing β cells. Although many experiments have shown promising results with stem cells in vitro, their clinical testing still needs more exploration. The review attempts to bring into light the clinical studies favoring the transplantation of stem cells in diabetic patients with an objective of improving insulin secretion and improving degeneration of different tissues in response to diabetes. It also focuses on the problems associated with successful implementation of the technique and possible directions for future research.
Collapse
Affiliation(s)
- Junaid Khan
- University Teaching Department (Pharmacy), Sarguja University, Ambikapur (Chhattisgarh) 497001, India
| | - Amit Alexander
- Rungta College of Pharmaceutical Sciences and Research, Bhilai, Chhattisgarh 490024, India
| | - Mukta Agrawal
- Rungta College of Pharmaceutical Sciences and Research, Bhilai, Chhattisgarh 490024, India
| | - Ajazuddin
- Rungta College of Pharmaceutical Sciences and Research, Bhilai, Chhattisgarh 490024, India
| | - Sunil Kumar Dubey
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Rajasthan, India
| | - Sabahuddin Siddique
- Patel College of Pharmacy, Madhyanchal Professional University, Bhopal, India
| | - Swarnlata Saraf
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur Chhattisgarh 492010, India
| | - Shailendra Saraf
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur Chhattisgarh 492010, India
| |
Collapse
|
24
|
Role of stem cell mobilization in the treatment of ischemic diseases. Arch Pharm Res 2019; 42:224-231. [PMID: 30680545 DOI: 10.1007/s12272-019-01123-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 01/16/2019] [Indexed: 12/13/2022]
Abstract
Stem cell mobilization plays important roles in the treatment of severe ischemic diseases, including myocardial infarction, limb ischemia, ischemic stroke, and acute kidney injury. Stem cell mobilization refers to the egress of heterogeneous stem cells residing in the bone marrow into the peripheral blood. In the clinic, granulocyte colony-stimulating factor (G-CSF) is the drug most commonly used to induce stem cell mobilization. Plerixafor, a direct antagonist of CXCR4, is also frequently used alone or in combination with G-CSF to mobilize stem cells. The molecular mechanisms by which G-CSF induces stem cell mobilization are well characterized. Briefly, G-CSF activates neutrophils in the bone marrow, which then release proteolytic enzymes, such as neutrophil elastase, cathepsin G, and matrix metalloproteinase 9, which cleave a variety of molecules responsible for stem cell retention in the bone marrow, including CXCL12, VCAM-1, and SCF. Subsequently, stem cells are released from the bone marrow into the peripheral blood. The released stem cells can be collected and used in autologous or allogeneic transplantation. To identify better conditions for stem cell mobilization in the treatment of acute and chronic ischemic diseases, several preclinical and clinical studies have been conducted over the past decade on various mobilizing agents. In this paper, we are going to review methods that induce mobilization of stem cells from the bone marrow and introduce the application of stem cell mobilization to therapy of ischemic diseases.
Collapse
|
25
|
Salybekov AA, Kawaguchi AT, Masuda H, Vorateera K, Okada C, Asahara T. Regeneration-associated cells improve recovery from myocardial infarction through enhanced vasculogenesis, anti-inflammation, and cardiomyogenesis. PLoS One 2018; 13:e0203244. [PMID: 30485279 PMCID: PMC6261405 DOI: 10.1371/journal.pone.0203244] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/23/2018] [Indexed: 11/18/2022] Open
Abstract
Background Considering the impaired function of regenerative cells in myocardial infarction (MI) patients with comorbidities and associated risk factors, cell therapy to enhance the regenerative microenvironment was designed using regeneration-associated cells (RACs), including endothelial progenitor cells (EPCs) and anti-inflammatory cells. Methods RACs were prepared by quality and quantity control culture of blood mononuclear cells (QQMNCs). Peripheral blood mononuclear cells (PBMNCs) were isolated from Lewis rats and conditioned for 5 days using a medium containing stem cell factors, thrombopoietin, Flt-3 ligand, vascular endothelial growth factor, and interleukin-6 to generate QQMNCs. Results A 5.3-fold increase in the definitive colony-forming EPCs and vasculogenic EPCs was observed, in comparison to naïve PBMNCs. QQMNCs were enriched with EPCs (28.9-fold, P<0.0019) and M2 macrophages (160.3-fold, P<0.0002). Genes involved in angiogenesis (angpt1, angpt2, and vegfb), stem/progenitors (c-kit and sca-1), and anti-inflammation (arg-1, erg-2, tgfb, and foxp3) were upregulated in QQMNCs. For in vivo experiments, cells were administered into syngeneic rat models of MI. QQMNC-transplanted group (QQ-Tx) preserved cardiac function and fraction shortening 28 days post-MI in comparison with PBMNCs-transplanted (PB-Tx) (P<0.0001) and Control (P<0.0008) groups. QQ-Tx showed enhanced angiogenesis and reduced interstitial left ventricular fibrosis, along with a decrease in neutrophils and an increase in M2 macrophages in the acute phase of MI. Cell tracing studies revealed that intravenously administered QQMNCs preferentially homed to ischemic tissues via blood circulation. QQ-Tx showed markedly upregulated early cardiac transcriptional cofactors (Nkx2-5, 29.8-fold, and Gata-4, 5.2-fold) as well as c-kit (4.5-fold) while these markers were downregulated in PB-Tx. In QQ-Tx animals, de novo blood vessels formed a “Biological Bypass”, observed macroscopically and microscopically, while PB-Tx and Control-Tx groups showed severe fibrotic adhesion to the surrounding tissues, but no epicardial blood vessels. Conclusion QQMNCs conferred potent angiogenic and anti-inflammatory properties to the regenerative microenvironment, enhancing myocardiogenesis and functional recovery of rat MI hearts.
Collapse
Affiliation(s)
- Amankeldi A. Salybekov
- Department of Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Japan
| | - Akira T. Kawaguchi
- Department of Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Japan
| | - Haruchika Masuda
- Department of Physiology, Tokai University School of Medicine, Isehara, Japan
| | - Kosit Vorateera
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok Noi, Thailand
| | - Chisa Okada
- Teaching and Research Support Core Center, Tokai University School of Medicine, Isehara, Japan
| | - Takayuki Asahara
- Department of Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Japan
- * E-mail:
| |
Collapse
|
26
|
Albiero M, Fadini GP. Pharmacologic targeting of the diabetic stem cell mobilopathy. Pharmacol Res 2018; 135:18-24. [PMID: 30030170 DOI: 10.1016/j.phrs.2018.07.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/26/2018] [Accepted: 07/16/2018] [Indexed: 01/01/2023]
Abstract
Diabetes is a chronic metabolic disease characterized by hyperglycemia and several associated biochemical abnormalities. Diabetes leads to multiorgan complications that collectively reduce life expectancy. Hematopoietic stem cells (HSCs) are nested within bone marrow (BM) niches whence they can be mobilized to the peripheral circulation. Clinically, this is done for HSC collection and autologous or allogenic transplantation. A great amount of data from basic and clinical studies support that diabetic patients are poor HSC mobilizers owing to BM remodeling. Dysfunction of the BM shares pathophysiological features and pathways with typical chronic diabetic complications that affect other issues (e.g. the retina and the kidney). From a clinical perspective, impaired HSC mobilization translates into the failure to collect a minimum number of CD34+ cells to achieve a safe engraftment after transplantation. Furthermore, blunted mobilization is associated with reduced steady-state levels of circulating HSCs, which have been consistently described in diabetic patients and associated with increased risk of adverse outcomes, including cardiovascular events and death. In this review, we discuss the most clinically relevant pharmacological options to overcome impaired HSC mobilization in diabetes. These therapeutic strategies may result in an improved outcome of diabetic patients undergoing HSC transplantation and restore circulating HSC levels, thereby protecting from adverse cardiovascular outcomes.
Collapse
Affiliation(s)
- M Albiero
- Venetian Institute of Molecular Medicine, Laboratory of Experimental Diabetology, 35100 Padova, Italy; Department of Medicine, Metabolic Division, University of Padova, 35100 Padova, Italy
| | - G P Fadini
- Venetian Institute of Molecular Medicine, Laboratory of Experimental Diabetology, 35100 Padova, Italy; Department of Medicine, Metabolic Division, University of Padova, 35100 Padova, Italy.
| |
Collapse
|
27
|
Wang B, Yu J, Wang T, Shen Y, Lin D, Xu X, Wang Y. Identification of megakaryocytes as a target of advanced glycation end products in diabetic complications in bone marrow. Acta Diabetol 2018; 55:419-427. [PMID: 29417230 DOI: 10.1007/s00592-018-1109-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 01/17/2018] [Indexed: 10/18/2022]
Abstract
AIMS To define the possible effect of diabetic conditions on megakaryocytes, the long-know precursors of platelets and lately characterized modulator of hematopoietic stem quiescence-activation transition. METHODS Megakaryoblastic MEG-01 cell culture and TPO/SCF/IL-3-induced differentiation of human umbilical blood mononuclear cells toward megakaryocytes were used to test effects of glycated bovine serum albumin (BSA-AGEs). The ob/ob mice and streptozotocin-treated mice were used as models of hyperglycemia. MTT was used to measure cell proliferation, FACS for surface marker and cell cycle, and RT-qPCR for the expression of interested genes. Megakaryocytes at different stages in marrow smear were checked under microscope. RESULTS When added in MEG-01 cultures at 200 μg/ml, BSA-AGEs increased proliferation of cells and enhanced mRNA expression of RAGE, VEGFα and PF4 in the cells. None of cell cycle distribution, PMA-induced platelet-like particles production, expression of GATA1/NF-E2/PU-1/IL-6/OPG/PDGF in MEG-01 cells nor TPO/SCF/IL-3 induced umbilical cord blood cells differentiation into megakaryocyte was affected by BSA-AGEs. In the ob/ob diabetic mice, MKs percentages in marrow cells and platelets in peripheral blood were significantly increased compared with control mice. In streptozotocin-induced diabetic mice, however, MKs percentage in marrow cells was decreased though peripheral platelet counts were not altered. Gene expression assay showed that the change in MKs in these two diabetic conditions might be explained by the alteration of GATA1 and NF-E2 expression, respectively. CONCLUSIONS Diabetic condition in animals might exert its influence on hematopoiesis via megakaryocytes-the newly identified modulator of hematopoietic stem cells in bone marrow.
Collapse
Affiliation(s)
- Benfang Wang
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 708 Renmin Road, Suzhou, 215007, China
| | - Jianjiang Yu
- Department of Clinical Laboratory, The Affiliated Jiangyin Hospital of Southeast University, Jiangyin, 214400, China
| | - Ting Wang
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 708 Renmin Road, Suzhou, 215007, China
| | - Ying Shen
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 708 Renmin Road, Suzhou, 215007, China
| | - Dandan Lin
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 708 Renmin Road, Suzhou, 215007, China
| | - Xin Xu
- Department of Hematology, The Affiliated Jiangyin Hospital of Southeast University, Jiangyin, 214400, China
| | - Yiqiang Wang
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 708 Renmin Road, Suzhou, 215007, China.
| |
Collapse
|
28
|
Siegel G, Fleck E, Elser S, Hermanutz-Klein U, Waidmann M, Northoff H, Seifried E, Schäfer R. Manufacture of endothelial colony-forming progenitor cells from steady-state peripheral blood leukapheresis using pooled human platelet lysate. Transfusion 2018; 58:1132-1142. [PMID: 29473177 DOI: 10.1111/trf.14541] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/20/2017] [Accepted: 12/23/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Endothelial colony-forming progenitor cells (ECFCs) are promising candidates for cell therapies. However, ECFC translation to the clinic requires optimized isolation and manufacture technologies according to good manufacturing practice (GMP). STUDY DESIGN AND METHODS ECFCs were manufactured from steady-state peripheral blood (PB) leukapheresis (11 donors), using GMP-compliant technologies including pooled human platelet (PLT) lysate, and compared to human umbilical cord endothelial cells, human aortic endothelial cells, and human cerebral microvascular endothelial cells. Specific variables assessed were growth kinetics, phenotype, trophic factors production, stimulation of tube formation, and Dil-AcLDL uptake. RESULTS ECFCs could be isolated from PB leukapheresis units with mean processed volume of 5411 mL and mean white blood cell (WBC) concentration factor of 8.74. The mean frequency was 1.44 × 10-8 ECFCs per WBC, corresponding to a mean of 177.8 ECFCs per apheresis unit. Expandable for up to 12 cumulative population doublings, calculated projection showed that approximately 730 × 103 ECFCs could be manufactured from 1 apheresis unit. ECFCs produced epidermal growth factor, hepatocyte growth factor, vascular endothelial growth factor (VEGF)-A, PLT-derived growth factor-B, interleukin-8, and monocyte chemoattractant protein-1, featured high potential for capillary-like tubes formation, and showed no telomerase activity. They were characterized by CD29, CD31, CD44, CD105, CD117, CD133, CD144, CD146, and VEGF-R2 expression, with the most common subpopulation CD34+CD117-CD133-. Compared to controls, ECFCs featured greater Dil-AcLDL uptake and higher expression of CD29, CD31, CD34, CD44, CD144, and VEGF-R2. CONCLUSIONS Here we show that isolation of ECFCs with proangiogenic profile from steady-state PB leukapheresis is feasible, marking a first step toward ECFC product manufacture according to GMP.
Collapse
Affiliation(s)
- Georg Siegel
- Institute for Clinical and Experimental Transfusion Medicine
| | - Erika Fleck
- Institute for Transfusion Medicine and Immunohaematology, German Red Cross Blood Donor Service Baden-Württemberg-Hessen gGmbH, Goethe-University Hospital, Frankfurt am Main, Germany
| | - Stefanie Elser
- Institute for Clinical and Experimental Transfusion Medicine.,Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, Tübingen, Germany
| | | | - Marc Waidmann
- Institute for Clinical and Experimental Transfusion Medicine
| | - Hinnak Northoff
- Institute for Clinical and Experimental Transfusion Medicine
| | - Erhard Seifried
- Institute for Transfusion Medicine and Immunohaematology, German Red Cross Blood Donor Service Baden-Württemberg-Hessen gGmbH, Goethe-University Hospital, Frankfurt am Main, Germany
| | - Richard Schäfer
- Institute for Clinical and Experimental Transfusion Medicine.,Institute for Transfusion Medicine and Immunohaematology, German Red Cross Blood Donor Service Baden-Württemberg-Hessen gGmbH, Goethe-University Hospital, Frankfurt am Main, Germany
| |
Collapse
|
29
|
Fadini GP, DiPersio JF. Diabetes mellitus as a poor mobilizer condition. Blood Rev 2017; 32:184-191. [PMID: 29132746 DOI: 10.1016/j.blre.2017.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 11/02/2017] [Accepted: 11/07/2017] [Indexed: 01/04/2023]
Abstract
Hematopoietic stem cell (HSC) transplantation in an effective and curative therapy for numerous hematological malignancies. Mobilization of HSCs from bone marrow (BM) to peripheral blood (PB) followed by apheresis is the gold standard for obtaining HSCs for both autologous and allogeneic stem cell transplantation. After administration of granulocyte-colony stimulating factor (G-CSF), up to 30% of patients fail to mobilize "optimal" numbers of HSCs required for engraftment. This review summarizes the current experimental and clinical evidence that diabetes mellitus is a risk factor for poor mobilization. Diabetes causes a profound remodeling of the HSC niche, resulting in impaired release of HSCs. Experimental studies indicate that hyperglycemia hampers regulation of CXCL12 and clinical studies suggest that diabetes impairs HSC mobilization especially in response to G-CSF, but less to plerixafor. Understanding further the biochemical alterations in the diabetic BM will provide insights into future therapeutic strategies to reverse the so-called "diabetic stem cell mobilopathy".
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine, 35128 Padova, Italy.
| | - John F DiPersio
- Washington University School of Medicine, St Louis, MO, United States.
| |
Collapse
|
30
|
Autologous Bone Marrow-Derived Stem Cells for Treating Diabetic Neuropathy in Metabolic Syndrome. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8945310. [PMID: 29098161 PMCID: PMC5643093 DOI: 10.1155/2017/8945310] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 08/23/2017] [Indexed: 01/08/2023]
Abstract
Diabetic neuropathy is one of the most common and serious complications of diabetes mellitus and metabolic syndrome. The current therapy strategies, including glucose control and pain management, are not effective for most patients. Growing evidence suggests that infiltration of inflammation factors and deficiency of local neurotrophic and angiogenic factors contribute significantly to the pathologies of diabetic neuropathy. Experimental and clinical studies have shown that bone marrow-derived stem cells (BMCs) therapy represents a novel and promising strategy for tissue repair through paracrine secretion of multiple cytokines, which has a potential to inhibit inflammation and promote angiogenesis and neurotrophy in diabetic neuropathy. In this review, we discuss the clinical practice in diabetic neuropathy and the therapeutic effect of BMC. We subsequently illustrate the functional impairment of autologous BMCs due to the interrupted bone marrow niche in diabetic neuropathy. We anticipate that the functional restoration of BMCs could improve their therapeutic effect and enable their wide applications in diabetic neuropathy.
Collapse
|
31
|
Abstract
Vascular complications contribute significantly to morbidity and mortality of diabetes mellitus. The primary cause of vascular complications in diabetes mellitus is hyperglycaemia, associated with endothelial dysfunction and impaired neovascularization. Circulating endothelial progenitor cells was shown to play important roles in vascular repair and promoting neovascularization. In this review, we will demonstrate the individual effect of high glucose on endothelial progenitor cells. Endothelial progenitor cells isolated from healthy subjects exposed to high glucose conditions or endothelial progenitor cells isolated from diabetic patients exhibit reduced number of endothelial cell colony forming units, impaired abilities of differentiation, proliferation, adhesion and migration, tubulization, secretion, mobilization and homing, whereas enhanced senescence. Increased production of reactive oxygen species by the mitochondria seems to play a crucial role in high glucose-induced endothelial progenitor cells deficit. Later, we will review the agents that might be used to alleviate dysfunction of endothelial progenitor cells induced by high glucose. The conclusions are that the relationship between hyperglycaemia and endothelial progenitor cells dysfunction is only beginning to be recognized, and future studies should pay more attention to the haemodynamic environment of endothelial progenitor cells and ageing factors to discover novel treatment agents.
Collapse
Affiliation(s)
- Hongyan Kang
- 1 Key Laboratory for Biomechanics and Mechanobiology of the Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Xuejiao Ma
- 1 Key Laboratory for Biomechanics and Mechanobiology of the Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Jiajia Liu
- 1 Key Laboratory for Biomechanics and Mechanobiology of the Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yubo Fan
- 1 Key Laboratory for Biomechanics and Mechanobiology of the Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- 2 National Research Center for Rehabilitation Technical Aids, Beijing, China
| | - Xiaoyan Deng
- 1 Key Laboratory for Biomechanics and Mechanobiology of the Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| |
Collapse
|
32
|
Satthenapalli VR, Lamberts RR, Katare RG. Concise Review: Challenges in Regenerating the Diabetic Heart: A Comprehensive Review. Stem Cells 2017; 35:2009-2026. [PMID: 28639375 DOI: 10.1002/stem.2661] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 06/07/2017] [Accepted: 06/13/2017] [Indexed: 12/15/2022]
Abstract
Stem cell therapy is one of the promising regenerative strategies developed to improve cardiac function in patients with ischemic heart diseases (IHD). However, this approach is limited in IHD patients with diabetes due to a progressive decline in the regenerative capacity of stem cells. This decline is mainly attributed to the metabolic memory incurred by diabetes on stem cell niche and their systemic cues. Understanding the molecular pathways involved in the diabetes-induced deterioration of stem cell function will be critical for developing new cardiac regeneration therapies. In this review, we first discuss the most common molecular alterations occurring in the diabetic stem cells/progenitor cells. Next, we highlight the key signaling pathways that can be dysregulated in a diabetic environment and impair the mobilization of stem/progenitor cells, which is essential for the transplanted/endogenous stem cells to reach the site of injury. We further discuss the possible methods of preconditioning the diabetic cardiac progenitor cell (CPC) with an aim to enrich the availability of efficient stem cells to regenerate the diseased diabetic heart. Finally, we propose new modalities for enriching the diabetic CPC through genetic or tissue engineering that would aid in developing autologous therapeutic strategies, improving the proliferative, angiogenic, and cardiogenic properties of diabetic stem/progenitor cells. Stem Cells 2017;35:2009-2026.
Collapse
Affiliation(s)
- Venkata R Satthenapalli
- Department of Physiology, School of Biomedical Sciences, HeartOtago, University of Otago, Dunedin, New Zealand
| | - Regis R Lamberts
- Department of Physiology, School of Biomedical Sciences, HeartOtago, University of Otago, Dunedin, New Zealand
| | - Rajesh G Katare
- Department of Physiology, School of Biomedical Sciences, HeartOtago, University of Otago, Dunedin, New Zealand
| |
Collapse
|
33
|
Abstract
Peripheral blood stem cell collection is an effective approach to obtain a hematopoietic graft for stem cell transplantation. Developing hematopoietic stem/progenitor cell (HSPC) mobilization methods and collection algorithms have improved efficiency, clinical outcomes, and cost effectiveness. Differences in mobilization mechanisms may change the HSPC content harvested and result in different engraftment kinetics and complications. Patient-specific factors can affect mobilization. Incorporating these factors in collection algorithms and improving assays for evaluating mobilization further extend the ability to obtain sufficient HSPCs for hematopoietic repopulation. Technological advance and innovations in leukapheresis have improved collection efficiency and reduced adverse effects.
Collapse
Affiliation(s)
- Yen-Michael S Hsu
- Pathology and Laboratory Medicine, Transfusion Medicine and Cellular Therapy, Weill Cornell Medical College, 525 East 68th Street, Box 251, New York, NY 10065, USA.
| | - Melissa M Cushing
- Transfusion Medicine and Cellular Therapy, Weill Cornell Medical College, 525 East 68th Street, Box 251, M09, New York, NY 10065, USA.
| |
Collapse
|
34
|
Park SS, Moisseiev E, Bauer G, Anderson JD, Grant MB, Zam A, Zawadzki RJ, Werner JS, Nolta JA. Advances in bone marrow stem cell therapy for retinal dysfunction. Prog Retin Eye Res 2017; 56:148-165. [PMID: 27784628 PMCID: PMC5237620 DOI: 10.1016/j.preteyeres.2016.10.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 10/11/2016] [Accepted: 10/18/2016] [Indexed: 12/21/2022]
Abstract
The most common cause of untreatable vision loss is dysfunction of the retina. Conditions, such as age-related macular degeneration, diabetic retinopathy and glaucoma remain leading causes of untreatable blindness worldwide. Various stem cell approaches are being explored for treatment of retinal regeneration. The rationale for using bone marrow stem cells to treat retinal dysfunction is based on preclinical evidence showing that bone marrow stem cells can rescue degenerating and ischemic retina. These stem cells have primarily paracrine trophic effects although some cells can directly incorporate into damaged tissue. Since the paracrine trophic effects can have regenerative effects on multiple cells in the retina, the use of this cell therapy is not limited to a particular retinal condition. Autologous bone marrow-derived stem cells are being explored in early clinical trials as therapy for various retinal conditions. These bone marrow stem cells include mesenchymal stem cells, mononuclear cells and CD34+ cells. Autologous therapy requires no systemic immunosuppression or donor matching. Intravitreal delivery of CD34+ cells and mononuclear cells appears to be tolerated and is being explored since some of these cells can home into the damaged retina after intravitreal administration. The safety of intravitreal delivery of mesenchymal stem cells has not been well established. This review provides an update of the current evidence in support of the use of bone marrow stem cells as treatment for retinal dysfunction. The potential limitations and complications of using certain forms of bone marrow stem cells as therapy are discussed. Future directions of research include methods to optimize the therapeutic potential of these stem cells, non-cellular alternatives using extracellular vesicles, and in vivo high-resolution retinal imaging to detect cellular changes in the retina following cell therapy.
Collapse
Affiliation(s)
- Susanna S Park
- Department of Ophthalmology & Vision Science, University of California Davis, Sacramento, CA, 95817, USA.
| | - Elad Moisseiev
- Department of Ophthalmology & Vision Science, University of California Davis, Sacramento, CA, 95817, USA.
| | - Gerhard Bauer
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis, Sacramento, CA, 95817, USA.
| | - Johnathon D Anderson
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis, Sacramento, CA, 95817, USA.
| | - Maria B Grant
- Department of Ophthalmology, Glick Eye Institute, Indiana University, Indianapolis, IN, USA.
| | - Azhar Zam
- UC Davis RISE Eye-Pod Small Animal Imaging Laboratory, Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA, USA.
| | - Robert J Zawadzki
- Department of Ophthalmology & Vision Science, University of California Davis, Sacramento, CA, 95817, USA; UC Davis RISE Eye-Pod Small Animal Imaging Laboratory, Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA, USA.
| | - John S Werner
- Department of Ophthalmology & Vision Science, University of California Davis, Sacramento, CA, 95817, USA.
| | - Jan A Nolta
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis, Sacramento, CA, 95817, USA.
| |
Collapse
|
35
|
Stocks BT, Thomas AB, Elizer SK, Zhu Y, Marshall AF, Wilson CS, Moore DJ. Hematopoietic Stem Cell Mobilization Is Necessary but Not Sufficient for Tolerance in Islet Transplantation. Diabetes 2017; 66:127-133. [PMID: 27797908 PMCID: PMC5204317 DOI: 10.2337/db16-0444] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 10/21/2016] [Indexed: 11/13/2022]
Abstract
Overcoming the immune response to establish durable immune tolerance in type 1 diabetes remains a substantial challenge. The ongoing effector immune response involves numerous immune cell types but is ultimately orchestrated and sustained by the hematopoietic stem cell (HSC) niche. We therefore hypothesized that tolerance induction also requires these pluripotent precursors. In this study, we determined that the tolerance-inducing agent anti-CD45RB induces HSC mobilization in nonautoimmune B6 mice but not in diabetes-prone NOD mice. Ablation of HSCs impaired tolerance to allogeneic islet transplants in B6 recipients. Mobilization of HSCs resulted in part from decreasing osteoblast expression of HSC retention factors. Furthermore, HSC mobilization required a functioning sympathetic nervous system; sympathectomy prevented HSC mobilization and completely abrogated tolerance induction. NOD HSCs were held in their niche by excess expression of CXCR4, which, when blocked, led to HSC mobilization and prolonged islet allograft survival. Overall, these findings indicate that the HSC compartment plays an underrecognized role in the establishment and maintenance of immune tolerance, and this role is disrupted in diabetes-prone NOD mice. Understanding the stem cell response to immune therapies in ongoing human clinical studies may help identify and maximize the effect of immune interventions for type 1 diabetes.
Collapse
Affiliation(s)
- Blair T Stocks
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Analise B Thomas
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University Medical Center, Nashville, TN
| | - Sydney K Elizer
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University Medical Center, Nashville, TN
| | - Yuantee Zhu
- Departartment of Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | - Andrew F Marshall
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University Medical Center, Nashville, TN
| | - Christopher S Wilson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Daniel J Moore
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
36
|
Wils J, Favre J, Bellien J. Modulating putative endothelial progenitor cells for the treatment of endothelial dysfunction and cardiovascular complications in diabetes. Pharmacol Ther 2016; 170:98-115. [PMID: 27773788 DOI: 10.1016/j.pharmthera.2016.10.014] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Diabetes induces a decrease in the number and function of different pro-angiogenic cell types generically designated as putative endothelial progenitor cells (EPC), which encompasses cells from myeloid origin that act in a paracrine fashion to promote angiogenesis and putative "true" EPC that contribute to endothelial replacement. This not only compromises neovasculogenesis in ischemic tissues but also impairs, at an early stage, the reendotheliziation process at sites of injury, contributing to the development of endothelial dysfunction and cardiovascular complications. Hyperglycemia, insulin resistance and dyslipidemia promote putative EPC dysregulation by affecting the SDF-1/CXCR-4 and NO pathways and the p53/SIRT1/p66Shc axis that contribute to their mobilization, migration, homing and vasculogenic properties. To optimize the clinical management of patients with hypoglycemic agents, statins and renin-angiotensin system inhibitors, which display pleiotropic effects on putative EPC, is a first step to improve their number and angiogenic potential but specific strategies are needed. Among them, mobilizing therapies based on G-CSF, erythropoietin or CXCR-4 antagonism have been developed to increase putative EPC number to treat ischemic diseases with or without prior cell isolation and transplantation. Growth factors, genetic and pharmacological strategies are also evaluated to improve ex vivo cultured EPC function before transplantation. Moreover, pharmacological agents increasing in vivo the bioavailability of NO and other endothelial factors demonstrated beneficial effects on neovascularization in diabetic ischemic models but their effects on endothelial dysfunction remain poorly evaluated. More experiments are warranted to develop orally available drugs and specific agents targeting p66Shc to reverse putative EPC dysfunction in the expected goal of preventing endothelial dysfunction and diabetic cardiovascular complications.
Collapse
Affiliation(s)
- Julien Wils
- Department of Pharmacology, Rouen University Hospital, Rouen, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1096, Rouen, France; University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Julie Favre
- MITOVASC Institute, Angers, France; Centre National de la Recherche Scientifique (CNRS) UMR 6214, Angers, France; INSERM U1083, Angers, France; University of Angers, Angers, France
| | - Jérémy Bellien
- Department of Pharmacology, Rouen University Hospital, Rouen, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1096, Rouen, France; University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France.
| |
Collapse
|
37
|
Lu XJ, Chen Q, Rong YJ, Yang GJ, Li CH, Xu NY, Yu CH, Wang HY, Zhang S, Shi YH, Chen J. LECT2 drives haematopoietic stem cell expansion and mobilization via regulating the macrophages and osteolineage cells. Nat Commun 2016; 7:12719. [PMID: 27596364 PMCID: PMC5025878 DOI: 10.1038/ncomms12719] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 07/26/2016] [Indexed: 12/12/2022] Open
Abstract
Haematopoietic stem cells (HSCs) can differentiate into cells of all lineages in the blood. However, the mechanisms by which cytokines in the blood affect HSC homeostasis remain largely unknown. Here we show that leukocyte cell-derived chemotaxin 2 (LECT2), a multifunctional cytokine, induces HSC expansion and mobilization. Recombinant LECT2 administration results in HSC expansion in the bone marrow and mobilization to the blood via CD209a. The effect of LECT2 on HSCs is reduced after specific depletion of macrophages or reduction of osteolineage cells. LECT2 treatment reduces the tumour necrosis factor (TNF) expression in macrophages and osteolineage cells. In TNF knockout mice, the effect of LECT2 on HSCs is reduced. Moreover, LECT2 induces HSC mobilization in irradiated mice, while granulocyte colony-stimulating factor does not. Our results illustrate that LECT2 is an extramedullar cytokine that contributes to HSC homeostasis and may be useful to induce HSC mobilization.
Collapse
Affiliation(s)
- Xin-Jiang Lu
- Laboratory of Biochemistry and Molecular Biology, Department of Marine and Life Sciences, Ningbo University, Ningbo 315211, China
| | - Qiang Chen
- Laboratory of Biochemistry and Molecular Biology, Department of Marine and Life Sciences, Ningbo University, Ningbo 315211, China
| | - Ye-Jing Rong
- Laboratory of Biochemistry and Molecular Biology, Department of Marine and Life Sciences, Ningbo University, Ningbo 315211, China
| | - Guan-Jun Yang
- Laboratory of Biochemistry and Molecular Biology, Department of Marine and Life Sciences, Ningbo University, Ningbo 315211, China
| | - Chang-Hong Li
- Laboratory of Biochemistry and Molecular Biology, Department of Marine and Life Sciences, Ningbo University, Ningbo 315211, China
| | - Ning-Yi Xu
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Chao-Hui Yu
- Department of Gastroenterology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Hui-Ying Wang
- Department of Allergy and Clinical Immunology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Shun Zhang
- Clinical Research Center, Ningbo No. 2 Hospital, Ningbo 315010, China
| | - Yu-Hong Shi
- Laboratory of Biochemistry and Molecular Biology, Department of Marine and Life Sciences, Ningbo University, Ningbo 315211, China
| | - Jiong Chen
- Laboratory of Biochemistry and Molecular Biology, Department of Marine and Life Sciences, Ningbo University, Ningbo 315211, China
| |
Collapse
|
38
|
Fadini GP, Ciciliot S, Albiero M. Concise Review: Perspectives and Clinical Implications of Bone Marrow and Circulating Stem Cell Defects in Diabetes. Stem Cells 2016; 35:106-116. [PMID: 27401837 DOI: 10.1002/stem.2445] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 05/27/2016] [Accepted: 05/28/2016] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus is a complex systemic disease characterized by severe morbidity and excess mortality. The burden of its multiorgan complications relies on an imbalance between hyperglycemic cell damage and defective endogenous reparative mechanisms. Inflammation and abnormalities in several hematopoietic components are typically found in diabetes. The discovery that diabetes reduces circulating stem/progenitor cells and impairs their function has opened an entire new field of study where diabetology comes into contact with hematology and regenerative medicine. It is being progressively recognized that such rare circulating cell populations mirror finely regulated processes involved in hematopoiesis, immunosurveillance, and peripheral tissue homeostasis. From a clinical perspective, pauperization of circulating stem cells predicts adverse outcomes and death. Furthermore, studies in murine models and humans have identified the bone marrow (BM) as a previously neglected site of diabetic end-organ damage, characterized by microangiopathy, neuropathy, fat deposition, and inflammation. As a result, diabetes impairs the mobilization of BM stem/progenitor cells, a defect known as mobilopathy or myelokathexis, with negative consequences for physiologic hematopoiesis, immune regulation, and tissue regeneration. A better understanding of the molecular and cellular processes that govern the BM stem cell niche, cell mobilization, and kinetics in peripheral tissues may uncover new therapeutic strategies for patients with diabetes. This concise review summarizes the current knowledge on the interplay between the BM, circulating stem cells, and diabetes, and sets the stages for future developments in the field. Stem Cells 2017;35:106-116.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, and Venetian Institute of Molecular Medicine, Padova, 35128, Italy
| | - Stefano Ciciliot
- Department of Medicine, University of Padova, and Venetian Institute of Molecular Medicine, Padova, 35128, Italy
| | - Mattia Albiero
- Department of Medicine, University of Padova, and Venetian Institute of Molecular Medicine, Padova, 35128, Italy
| |
Collapse
|
39
|
Ben Nasr M, Fiorina P. CXCR4 antagonism overcomes diabetic stem cell mobilopathy. Atherosclerosis 2016; 251:512-513. [PMID: 27352994 DOI: 10.1016/j.atherosclerosis.2016.06.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 06/10/2016] [Indexed: 12/27/2022]
Affiliation(s)
- Moufida Ben Nasr
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Transplant Medicine, San Raffaele Hospital, Milan, Italy
| | - Paolo Fiorina
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Transplant Medicine, San Raffaele Hospital, Milan, Italy.
| |
Collapse
|
40
|
Fiala MA, Slade M, Wang S, Park S, DiPersio J, Stockerl-Goldstein K. The impact of diabetes mellitus and other comorbidities on hematopoietic stem cell collection and hematologic recovery post-transplantation. Leuk Lymphoma 2016; 58:241-243. [PMID: 27243112 DOI: 10.1080/10428194.2016.1185788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Mark A Fiala
- a Division of Oncology, Department of Medicine , Washington University School of Medicine , St. Louis , MO , USA
| | - Michael Slade
- a Division of Oncology, Department of Medicine , Washington University School of Medicine , St. Louis , MO , USA
| | - Stephanie Wang
- a Division of Oncology, Department of Medicine , Washington University School of Medicine , St. Louis , MO , USA
| | - Soo Park
- a Division of Oncology, Department of Medicine , Washington University School of Medicine , St. Louis , MO , USA
| | - John DiPersio
- a Division of Oncology, Department of Medicine , Washington University School of Medicine , St. Louis , MO , USA
| | - Keith Stockerl-Goldstein
- a Division of Oncology, Department of Medicine , Washington University School of Medicine , St. Louis , MO , USA
| |
Collapse
|
41
|
Cappellari R, D'Anna M, Avogaro A, Fadini GP. Plerixafor improves the endothelial health balance. The effect of diabetes analysed by polychromatic flow cytometry. Atherosclerosis 2016; 251:373-380. [PMID: 27255499 DOI: 10.1016/j.atherosclerosis.2016.05.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/03/2016] [Accepted: 05/18/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND AIMS Diabetes damages the endothelium and reduces the availability of bone marrow (BM)-derived endothelial progenitor cells (EPCs). The mobilization of hematopoietic stem cells (HSCs) and EPCs in response to G-CSF is impaired by diabetes, owing to CXCL12 dysregulation. We have previously shown that the CXCR4/CXCL12 disruptor plerixafor rescues HSC and EPC mobilization in diabetes. We herein explored the effects of plerixafor on HSCs, EPCs, and circulating endothelial cells (CECs) in patients with and without diabetes. METHODS We re-analysed data gathered in the NCT02056210 trial, wherein patients with (n = 10) and without diabetes (n = 10) received plerixafor to test stem/progenitor cell mobilization. We applied a novel and very specific polychromatic flow cytometry (PFC) approach to identify and quantify HSCs, EPCs, and CECs. RESULTS We found that 7-AAD(-)Syto16(+)CD34(+)CD45(dim) HSC levels determined by PFC strongly correlated to the traditional enumeration of CD34(+) cells, whereas 7-AAD(-)Syto16(+)CD34(+)CD45(neg)KDR(+) EPCs were unrelated to the traditional enumeration of CD34(+)KDR(+) cells. Using PFC, we confirmed that plerixafor induces rapid mobilization of HSCs and EPCs in both groups, with a marginally significant defect in patients with diabetes. Plerixafor reduced live (7-AAD(-)) and dead (7-AAD(+)) Syto16(+)CD34(bright)CD45(neg)CD146(+) CECs more in patients without than in those with diabetes. The EPC/CEC ratio, a measure of the vascular health balance, was increased by plerixafor, but less prominently in patients with that in those without diabetes. CONCLUSIONS In addition to rescuing defective mobilization associated with diabetes, plerixafor improves the balance between EPCs and CECs, but the latter effect is blunted in patients with diabetes.
Collapse
Affiliation(s)
- Roberta Cappellari
- Department of Medicine, University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine, 35128 Padova, Italy
| | - Marianna D'Anna
- Department of Medicine, University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine, 35128 Padova, Italy
| | - Angelo Avogaro
- Department of Medicine, University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine, 35128 Padova, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine, 35128 Padova, Italy.
| |
Collapse
|
42
|
Abstract
Diabetes is one of the main economic burdens in health care, which threatens to worsen dramatically if prevalence forecasts are correct. What makes diabetes harmful is the multi-organ distribution of its microvascular and macrovascular complications. Regenerative medicine with cellular therapy could be the dam against life-threatening or life-altering complications. Bone marrow-derived stem cells are putative candidates to achieve this goal. Unfortunately, the bone marrow itself is affected by diabetes, as it can develop a microangiopathy and neuropathy similar to other body tissues. Neuropathy leads to impaired stem cell mobilization from marrow, the so-called mobilopathy. Here, we review the role of bone marrow-derived stem cells in diabetes: how they are affected by compromised bone marrow integrity, how they contribute to other diabetic complications, and how they can be used as a treatment for these. Eventually, we suggest new tactics to optimize stem cell therapy.
Collapse
Affiliation(s)
- Giuseppe Mangialardi
- Bristol Heart Institute, University of Bristol, Level 7, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS28HW UK
| | - Paolo Madeddu
- Bristol Heart Institute, University of Bristol, Level 7, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS28HW UK
| |
Collapse
|
43
|
Chong MSK, Ng WK, Chan JKY. Concise Review: Endothelial Progenitor Cells in Regenerative Medicine: Applications and Challenges. Stem Cells Transl Med 2016; 5:530-8. [PMID: 26956207 DOI: 10.5966/sctm.2015-0227] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 12/07/2015] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Endothelial progenitor cells (EPCs) are currently being studied as candidate cell sources for revascularization strategies. Significant advances have been made in understanding the biology of EPCs, and preclinical studies have demonstrated the vasculogenic, angiogenic, and beneficial paracrine effects of transplanted EPCs in the treatment of ischemic diseases. Despite these promising results, widespread clinical acceptance of EPCs for clinical therapies remains hampered by several challenges. The present study provides a concise summary of the different EPC populations being studied for ischemic therapies and their known roles in the healing of ischemic tissues. The challenges and issues surrounding the use of EPCs and the current strategies being developed to improve the harvest efficiency and functionality of EPCs for application in regenerative medicine are discussed. SIGNIFICANCE Endothelial progenitor cells (EPCs) have immense clinical value for cardiovascular therapies. The present study provides a concise description of the EPC subpopulations being evaluated for clinical applications. The current major lines of investigation involving preclinical and clinical evaluations of EPCs are discussed, and significant gaps limiting the translation of EPCs are highlighted. The present report could be useful for clinicians and clinical researchers with interests in ischemic therapy and for basic scientists working in the related fields of tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Mark Seow Khoon Chong
- School of Chemical and Biochemical Engineering, Nanyang Technological University, Singapore
| | - Wei Kai Ng
- School of Chemical and Biochemical Engineering, Nanyang Technological University, Singapore
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore Department of Obstetrics and Gynaecology, National University of Singapore, Singapore
| |
Collapse
|
44
|
Yang JZ, Zhang JQ, Sun LX. Mechanisms for T cell tolerance induced with granulocyte colony-stimulating factor. Mol Immunol 2015; 70:56-62. [PMID: 26703218 DOI: 10.1016/j.molimm.2015.12.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/02/2015] [Accepted: 12/03/2015] [Indexed: 12/15/2022]
Abstract
Granulocyte colony-stimulating factor (G-CSF) has been widely accepted as a mediator of T cell tolerance. The immune modulatory effect of G-CSF on T cells is believed to be mediated exclusively through other effector cells, such as monocytes, tolerogenic dendritic cells (DC), and myeloid-derived suppressor cells. Recent advances confirmed the direct effects of G-CSF in inducing immune tolerance of T cells through the G-CSF-G-CSF receptor pathway and related molecular mechanisms. This review aims to summarize the findings associated with the direct and indirect mechanisms for T cell tolerance induced with G-CSF. The role of G-CSF in preventing graft-versus-host disease (GVHD) and in treating autoimmune diseases (ADs) is also discussed. It is conceivable that G-CSF and immune cell compositions, such as tolerogenic DC and CD4(+)CD25(+)Foxp3(+) T cells, modulated by G-CSF could become an integral part of the immunomodulatory therapies against GVHD and ADs in the future.
Collapse
Affiliation(s)
- Jian-Zhu Yang
- Department of Pathology, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jin-Qiao Zhang
- Department of Hematology, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Li-Xia Sun
- Department of Hematology, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
45
|
Haller MJ, Atkinson MA, Wasserfall CH, Brusko TM, Mathews CE, Hulme M, Cintron M, Shuster J, McGrail K, Posgai A, Schatz D. Mobilization without immune depletion fails to restore immunological tolerance or preserve beta cell function in recent onset type 1 diabetes. Clin Exp Immunol 2015; 183:350-7. [PMID: 26462724 DOI: 10.1111/cei.12731] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2015] [Indexed: 12/12/2022] Open
Abstract
Granulocyte colony-stimulating factor (G-CSF) has been used to restore immune competence following chemoablative cancer therapy and to promote immunological tolerance in certain settings of autoimmunity. Therefore, we tested the potential of G-CSF to impact type 1 diabetes (T1D) progression in patients with recent-onset disease [n = 14; n = 7 (placebo)] and assessed safety, efficacy and mechanistic effects on the immune system. We hypothesized that pegylated G-CSF (6 mg administered subcutaneously every 2 weeks for 12 weeks) would promote regulatory T cell (Treg) mobilization to a degree capable of restoring immunological tolerance, thus preventing further decline in C-peptide production. Although treatment was well tolerated, G-CSF monotherapy did not affect C-peptide production, glycated haemoglobin (HbA1c) or insulin dose. Mechanistically, G-CSF treatment increased circulating neutrophils during the 12-week course of therapy (P < 0·01) but did not alter Treg frequencies. No effects were observed for CD4(+) : CD8(+) T cell ratio or the ratio of naive : memory (CD45RA(+)/CD45RO(+)) CD4(+) T cells. As expected, manageable bone pain was common in subjects receiving G-CSF, but notably, no severe adverse events such as splenomegaly occurred. This study supports the continued exploration of G-CSF and other mobilizing agents in subjects with T1D, but only when combined with immunodepleting agents where synergistic mechanisms of action have previously demonstrated efficacy towards the preservation of C-peptide.
Collapse
Affiliation(s)
- M J Haller
- Departments of Pediatric Endocrinology, University of Florida, Gainesville, FL, USA
| | - M A Atkinson
- Departments of Pediatric Endocrinology, University of Florida, Gainesville, FL, USA.,Immunology, Pathology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - C H Wasserfall
- Immunology, Pathology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - T M Brusko
- Immunology, Pathology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - C E Mathews
- Immunology, Pathology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - M Hulme
- Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - M Cintron
- Departments of Pediatric Endocrinology, University of Florida, Gainesville, FL, USA
| | - J Shuster
- Health Outcomes and Policy, University of Florida, Gainesville, FL, USA
| | - K McGrail
- Immunology, Pathology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - A Posgai
- Immunology, Pathology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - D Schatz
- Departments of Pediatric Endocrinology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
46
|
Spinetti G, Mangialardi G, Specchia C, Madeddu P. Enhancing Stem Cell Mobility: New Hope for Treatment of Cardiovascular Complications in Patients With Diabetes? Diabetes 2015. [PMID: 26207034 DOI: 10.2337/db15-0433] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | | | - Claudia Specchia
- IRCCS MultiMedica, Milan, Italy University of Brescia, Brescia, Italy
| | - Paolo Madeddu
- Bristol Heart Institute, University of Bristol, Bristol, U.K.
| |
Collapse
|
47
|
|