1
|
Scapin G, Cagdas E, Grav LM, Lewis NE, Goletz S, Hafkenscheid L. Implications of glycosylation for the development of selected cytokines and their derivatives for medical use. Biotechnol Adv 2024; 77:108467. [PMID: 39447666 DOI: 10.1016/j.biotechadv.2024.108467] [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: 03/06/2024] [Revised: 09/13/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024]
Abstract
Cytokines are important regulators of immune responses, making them attractive targets for autoimmune diseases and cancer therapeutics. Yet, the significance of cytokine glycosylation remains underestimated. Many cytokines carry N- and O-glycans and some even undergo C-mannosylation. Recombinant cytokines produced in heterologous host cells may lack glycans or exhibit a different glycosylation pattern such as varying levels of galactosylation, sialylation, fucosylation or xylose addition compared to their human counterparts, potentially impacting critical immune interactions. We focused on cytokines that are currently utilized or designed in advanced therapeutic formats, including immunocytokines, fusokines, engager cytokines, and genetically engineered 'supercytokines.' Despite the innovative designs of these cytokine derivatives, their glycosylation patterns have not been extensively studied. By examining the glycosylation of the human native cytokines, G-CSF and GM-CSF, interferons β and γ, TNF-α and interleukins-2, -3 -4, -6, -7, -9, -12, -13, -15, -17A, -21, and - 22, we aim to assess its potential impact on their therapeutic derivatives. Understanding the glycosylation of the native cytokines could provide critical insights into the safety, efficacy, and functionality of these next-generation cytokine therapies, affecting factors such as stability, bioactivity, antigenicity, and half-life. This knowledge can guide the choice of optimal expression hosts for production and advance the development of effective cytokine-based therapeutics and synthetic immunology drugs.
Collapse
Affiliation(s)
- Giulia Scapin
- Department of Biotechnology and Biomedicine, Mammalian Cell Line Engineering, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark
| | - Ece Cagdas
- Department of Biotechnology and Biomedicine, Mammalian Cell Line Engineering, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark
| | - Lise Marie Grav
- Department of Biotechnology and Biomedicine, Mammalian Cell Line Engineering, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark; The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Steffen Goletz
- Department of Biotechnology and Biomedicine, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark.
| | - Lise Hafkenscheid
- Department of Biotechnology and Biomedicine, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark.
| |
Collapse
|
2
|
Nikolaev B, Yakovleva L, Fedorov V, Yudintceva N, Ryzhov V, Marchenko Y, Ischenko A, Zhakhov A, Dobrodumov A, Combs SE, Gao H, Shevtsov M. Magnetic Relaxation Switching Assay Using IFNα-2b-Conjugated Superparamagnetic Nanoparticles for Anti-Interferon Antibody Detection. BIOSENSORS 2023; 13:624. [PMID: 37366989 DOI: 10.3390/bios13060624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/21/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023]
Abstract
Type I interferons, particularly IFNα-2b, play essential roles in eliciting adaptive and innate immune responses, being implicated in the pathogenesis of various diseases, including cancer, and autoimmune and infectious diseases. Therefore, the development of a highly sensitive platform for analysis of either IFNα-2b or anti-IFNα-2b antibodies is of high importance to improve the diagnosis of various pathologies associated with the IFNα-2b disbalance. For evaluation of the anti-IFNα-2b antibody level, we have synthesized superparamagnetic iron oxide nanoparticles (SPIONs) coupled with the recombinant human IFNα-2b protein (SPIONs@IFNα-2b). Employing a magnetic relaxation switching assay (MRSw)-based nanosensor, we detected picomolar concentrations (0.36 pg/mL) of anti-INFα-2b antibodies. The high sensitivity of the real-time antibodies' detection was ensured by the specificity of immune responses and the maintenance of resonance conditions for water spins by choosing a high-frequency filling of short radio-frequency pulses of the generator. The formation of a complex of the SPIONs@IFNα-2b nanoparticles with the anti-INFα-2b antibodies led to a cascade process of the formation of nanoparticle clusters, which was further enhanced by exposure to a strong (7.1 T) homogenous magnetic field. Obtained magnetic conjugates exhibited high negative MR contrast-enhancing properties (as shown by NMR studies) that were also preserved when particles were administered in vivo. Thus, we observed a 1.2-fold decrease of the T2 relaxation time in the liver following administration of magnetic conjugates as compared to the control. In conclusion, the developed MRSw assay based on SPIONs@IFNα-2b nanoparticles represents an alternative immunological probe for the estimation of anti-IFNα-2b antibodies that could be further employed in clinical studies.
Collapse
Affiliation(s)
- Boris Nikolaev
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), Tikhoretsky Ave., 4, 194064 St. Petersburg, Russia
| | - Ludmila Yakovleva
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), Tikhoretsky Ave., 4, 194064 St. Petersburg, Russia
| | - Viacheslav Fedorov
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), Tikhoretsky Ave., 4, 194064 St. Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, Akkuratova Str. 2, 197341 St. Petersburg, Russia
- Department of Inorganic Chemistry and Biophysics, Saint-Petersburg State University of Veterinary Medicine, Chernigovskaya Str. 5, 196084 St. Petersburg, Russia
| | - Natalia Yudintceva
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), Tikhoretsky Ave., 4, 194064 St. Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, Akkuratova Str. 2, 197341 St. Petersburg, Russia
| | - Vyacheslav Ryzhov
- Petersburg Nuclear Physics Institute, National Research Centre "Kurchatov Institute", 188300 Gatchina, Russia
| | - Yaroslav Marchenko
- Petersburg Nuclear Physics Institute, National Research Centre "Kurchatov Institute", 188300 Gatchina, Russia
| | - Alexander Ischenko
- Laboratory of Hybridoma Technologies, Saint-Petersburg Pasteur Institute, Mira Str. 14, 197101 St. Petersburg, Russia
| | - Alexander Zhakhov
- Laboratory of Hybridoma Technologies, Saint-Petersburg Pasteur Institute, Mira Str. 14, 197101 St. Petersburg, Russia
| | - Anatoliy Dobrodumov
- Department of Nuclear Magnetic Resonance, Institute of Macromolecular Compounds of the Russian Academy of Sciences (RAS), Bolshoi pr. 31, 199004 St. Petersburg, Russia
| | - Stephanie E Combs
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum Rechts der Isar, Ismaninger Str. 22, 81675 Munich, Germany
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Maxim Shevtsov
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), Tikhoretsky Ave., 4, 194064 St. Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, Akkuratova Str. 2, 197341 St. Petersburg, Russia
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum Rechts der Isar, Ismaninger Str. 22, 81675 Munich, Germany
- Laboratory of Biomedical Cell Technologies, Far Eastern Federal University, 690091 Vladivostok, Russia
| |
Collapse
|
3
|
Albiero M, D'Anna M, Bonora BM, Zuccolotto G, Rosato A, Giorgio M, Iori E, Avogaro A, Fadini GP. Hematopoietic and Nonhematopoietic p66Shc Differentially Regulates Stem Cell Traffic and Vascular Response to Ischemia in Diabetes. Antioxid Redox Signal 2022; 36:593-607. [PMID: 34538132 DOI: 10.1089/ars.2021.0097] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Aims: Peripheral artery disease (PAD) is a severe complication of diabetes, characterized by defective traffic of hematopoietic stem/progenitor cells (HSPCs). We examined the hematopoietic versus nonhematopoietic role of p66Shc in regulating HSPC traffic and blood flow recovery after ischemia in diabetic mice. Results: Using streptozotocin-induced diabetes, chimeric mice with green fluorescent protein (GFP)+ bone marrow (BM), and the hind limb ischemia model, we found that the physiologic mobilization and homing of HSPCs were abolished by diabetes, along with impaired vascular recovery. Hematopoietic deletion of p66Shc, obtained by transplanting p66Shc-/- BM cells into wild-type (Wt) recipients, but not nonhematopoietic deletion, constrained hyperglycemia-induced myelopoiesis, rescued postischemic HSPC mobilization, and improved blood flow recovery in diabetic mice. In Wt diabetic mice transplanted with BM cells from GFP+p66Shc-/- mice, the amount of HSPCs homed to ischemic muscles was greater than in mice transplanted with GFP+p66Shc+/+ cells, with recruited cells displaying higher expression of adhesion molecules and Vegf. In 40 patients with diabetes, p66Shc gene expression in mononuclear cells was correlated with myelopoiesis and elevated in the presence of PAD. In 13 patients with diabetes and PAD, p66Shc expression in HSPC-mobilized peripheral blood cells was inversely correlated with VEGF expression. Innovation: For the first time, we dissect the role of hematopoietic versus nonhematopoietic p66Shc in regulating HSPC traffic and ischemic responses. Conclusion: Hematopoietic deletion of p66Shc was sufficient to rescue HSPC mobilization and homing in diabetes after ischemia and improved blood flow recovery. Inhibiting p66Shc in blood cells may be a novel strategy to counter PAD in diabetes. Antioxid. Redox Signal. 36, 593-607. Clinical Trial No.: NCT02790957.
Collapse
Affiliation(s)
- Mattia Albiero
- Department of Medicine, University of Padova, Padova, Italy.,Veneto Institute of Molecular Medicine, Padova, Italy
| | - Marianna D'Anna
- Department of Medicine, University of Padova, Padova, Italy.,Veneto Institute of Molecular Medicine, Padova, Italy
| | - Benedetta Maria Bonora
- Department of Medicine, University of Padova, Padova, Italy.,Veneto Institute of Molecular Medicine, Padova, Italy
| | - Gaia Zuccolotto
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Padova, Italy
| | - Antonio Rosato
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Padova, Italy.,Veneto Institute of Oncology - IOV IRCCS, Padua, Italy
| | - Marco Giorgio
- European Institute of Oncology (IEO), Milan, Italy.,Department of Biomedical 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
| |
Collapse
|
4
|
Asialoglycoprotein receptor expression in placenta of women with Hepatitis B Virus e Antigen (HBeAg) positive and negative. ENFERMERIA CLINICA 2021. [PMID: 32204159 DOI: 10.1016/j.enfcli.2019.10.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE This study aimed to determine the relationship between Asialoglycoprotein receptor (ASGP-R) on the placenta and positivity of HBeAg in the mother's serum. METHOD We collected 52 placentas from delivered mothers who have HbsAg-positive serum. The HbsAg-positive serum was then examined for HBeAg-positive and HBeAg-negative. Immunohistochemistry staining was performed on block paraffin sections using monoclonal antibody of ASGP-R. RESULTS The expression of ASGP-R of 52 placenta samples demonstrated that 37 samples were scored I, five samples were score II, two samples were score III, and eight samples were score IV. We found that 14 of 52 serum samples were HBeAg-positive and 38 were HBeAg-negative. CONCLUSION There is a significant correlation between ASGP-R on the placenta and positivity of HBeAg in the mother's serum. The expression of the ASGP-R could increase the risk of HBV transmission. The result of this study could be used as a guideline for preventing and therapeutic approach of HBV from mother to child.
Collapse
|
5
|
Shimizu Y, Yamanashi H, Noguchi Y, Koyamatsu J, Nagayoshi M, Kiyoura K, Fukui S, Tamai M, Kawashiri SY, Kondo H, Maeda T. Cardio-ankle vascular index and circulating CD34-positive cell levels as indicators of endothelial repair activity in older Japanese men. Geriatr Gerontol Int 2019; 19:557-562. [PMID: 30920121 DOI: 10.1111/ggi.13657] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 02/13/2019] [Accepted: 02/20/2019] [Indexed: 02/02/2023]
Abstract
AIM The cardio-ankle vascular index (CAVI) reflects functional arterial stiffness, which is related to endothelial dysfunction. CD34-positive cells carry out an important function in endothelial repair. However, there have been no reports assessing the association between CAVI and the number of circulating CD34-positive cells. METHODS We carried out a cross-sectional study of 249 Japanese men, aged 60-69 years, who underwent annual health checkups between 2013 and 2015. As individuals with high levels of circulating CD34-positive cells might indicate the influence of consumptive reduction of circulating CD34-positive cells as a result of aggressive endothelial repair, participants were stratified by circulating CD34-positive cell levels, using the median value in this population (0.95 cells/μL) as the cut-off. RESULTS For participants with low circulating CD34-positive cell levels, logarithmic values of circulating CD34-positive cells were inversely associated with CAVI (multivariable standardized parameter estimate [β] = -0.22, P = 0.014), but not for participants with high levels (β = -0.04, P = 0.638). In addition, even when no significant associations between CAVI and carotid intima-media thickness were detected for participants with low circulating CD34-positive cell levels (β = -0.02, P = 0.865), significant positive associations were identified for participants with high levels (β = 0.22, P = 0.028). CONCLUSIONS As circulating CD34-positive cell count might indicate endothelial repair activity, the present results show that CAVI is affected by insufficient endothelial repair in individuals with low circulating CD34-positive cell counts. Our results also show that a positive association between CAVI and carotid intima-media thickness exists only in individuals with aggressive endothelial repair, which indicates the presence of organic arterial disease, such as atherosclerosis. Geriatr Gerontol Int 2019; 19: 557-562.
Collapse
Affiliation(s)
- Yuji Shimizu
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of Cardiovascular Disease Prevention, Osaka Center for Cancer and Cardiovascular Disease Prevention, Osaka, Japan
| | - Hirotomo Yamanashi
- Department of General Medicine, Nagasaki University Hospital, Nagasaki, Japan
| | - Yuko Noguchi
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Jun Koyamatsu
- Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Mako Nagayoshi
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kairi Kiyoura
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shoichi Fukui
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Mami Tamai
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shin-Ya Kawashiri
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hideaki Kondo
- Department of General Medicine, Nagasaki University Hospital, Nagasaki, Japan
| | - Takahiro Maeda
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of General Medicine, Nagasaki University Hospital, Nagasaki, Japan.,Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
6
|
Increased Endothelial Progenitor Cell Number in Early Stage of Endometrial Cancer. Int J Gynecol Cancer 2018; 27:947-952. [PMID: 28498245 DOI: 10.1097/igc.0000000000000961] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES It is generally believed that circulating endothelial cells (CECs) and endothelial progenitor cells (EPCs) reflect the state of the endothelium, its injury and/or repair possibilities. In different types of cancers, increased numbers of CECs and EPCs were found, suggesting their participation in cancer angiogenesis. The objective of this study was to determine whether, in the blood circulation of women with early endometrial cancer, CEC and EPC levels differ from those of healthy women of similar age. METHODS For CEC number evaluation, samples of peripheral blood cells of women with endometrial carcinoma and control subjects were labeled with anti-CD31 and anti-CD45 antibodies; for EPCs, with anti-VEGFR2 (vascular-endothelium growth factor receptor 2)/KDR and anti-CD34 antibodies. The CEC and EPC cells were then quantified by flow cytometry. RESULTS Endothelial progenitor cell numbers (CD34, VEGFR2/KDR) in the peripheral blood of women with endometrial carcinoma were significantly augmented as compared with those of control healthy women and CEC numbers (CD31, CD45) were similar in both groups. Cancer patients were divided according to the grading into G1 and G2 groups and according to the stage into International Federation of Gynecology and Obstetrics (FIGO) stage IA and FIGO IB groups. Statistically significant augmented EPC numbers were demonstrated only in G1 and FIGO IA patients. CONCLUSIONS These results strongly suggest new vessel formation from recruited endothelial precursors as being involved mainly at the early stages of tumor progression.
Collapse
|
7
|
Shimizu Y, Sato S, Noguchi Y, Koyamatsu J, Yamanashi H, Nagayoshi M, Kadota K, Kawashiri SY, Nagata Y, Maeda T. Triglycerides and blood pressure in relation to circulating CD34-positive cell levels among community-dwelling elderly Japanese men: a cross-sectional study. Environ Health Prev Med 2017; 22:77. [PMID: 29165175 PMCID: PMC5698944 DOI: 10.1186/s12199-017-0684-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/09/2017] [Indexed: 01/20/2023] Open
Abstract
Background Triglycerides are reported to be positively associated with blood pressure (both systolic and diastolic). However, in a previous study, we reported a significant positive association between triglycerides and circulating CD34-positive cells (endothelial repair) among non-hypertensive, but not hypertensive, participants. Since hypertension and endothelial dysfunction have a bi-directional association (vicious cycle), the status of circulating CD34-positive cells may influence the association between triglycerides and hypertension. Methods Since antihypertensive medication use may influence results of the present study, we conducted a cross-sectional study of 327 community dwelling elderly (aged 60–69 years) Japanese participants who were not taking anti-hypertensive medication and who had participated in a general health check-up in 2013–2015. Results Participants were classified into two groups based on median values of circulating CD34-positive cells (0.93 cells/μL). For participants with lower circulating CD34-positive cells (n = 165), a significant positive association was seen between triglycerides and blood pressure, but not for participants with higher circulating CD34-positive cells (n = 162). The multivariable standardized parameter estimates (β) and p values of systolic blood pressure and diastolic blood pressure were 0.23 (p = 0.007) and 0.18 (p = 0.036) for participants with lower circulating CD34-positive cells and 0.08 (p = 0.409) and 0.03 (p = 0.786) for those with higher circulating CD34-positive cells. Conclusion A significant positive association between triglycerides and blood pressure exists among those with lower, but not higher, circulating CD34-positive cells. The level of circulating CD34-positive cells acts as a determinant factor for the association between triglycerides and blood pressure.
Collapse
Affiliation(s)
- Yuji Shimizu
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Sakamoto 1-12-4, Nagasaki, 852-8523, Japan. .,Department of Cardiovascular Disease Prevention, Osaka Center for Cancer and Cardiovascular Disease Prevention, Osaka, Japan.
| | - Shimpei Sato
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Sakamoto 1-12-4, Nagasaki, 852-8523, Japan
| | - Yuko Noguchi
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Sakamoto 1-12-4, Nagasaki, 852-8523, Japan
| | - Jun Koyamatsu
- Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hirotomo Yamanashi
- Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Mako Nagayoshi
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Sakamoto 1-12-4, Nagasaki, 852-8523, Japan
| | - Koichiro Kadota
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Sakamoto 1-12-4, Nagasaki, 852-8523, Japan
| | - Shin-Ya Kawashiri
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Sakamoto 1-12-4, Nagasaki, 852-8523, Japan
| | - Yasuhiro Nagata
- Center for Comprehensive Community Care Education, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takahiro Maeda
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Sakamoto 1-12-4, Nagasaki, 852-8523, Japan.,Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
8
|
Fujita Y, Kawamoto A. Stem cell-based peripheral vascular regeneration. Adv Drug Deliv Rev 2017; 120:25-40. [PMID: 28912015 DOI: 10.1016/j.addr.2017.09.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/28/2017] [Accepted: 09/07/2017] [Indexed: 02/07/2023]
Abstract
Chronic critical limb ischemia (CLI) represents an end-stage manifestation of peripheral arterial disease (PAD). CLI patients are at very high risk of amputation and cardiovascular complications, leading to severe morbidity and mortality. Because many patients with CLI are ineligible for conventional revascularization procedures, it is urgently needed to explore alternative strategies to improve blood supply in the ischemic tissue. Although researchers initially focused on gene/protein therapy using proangiogenic growth factors/cytokines, recent discovery of somatic stem/progenitor cells including bone marrow (BM)-derived endothelial progenitor cells (EPCs) and mesenchymal stem cells (MSCs) has drastically developed the field of therapeutic angiogenesis for CLI. Overall, early phase clinical trials demonstrated that stem/progenitor cell therapies may be safe, feasible and potentially effective. However, only few late-phase clinical trials have been conducted. This review provides an overview of the preclinical and clinical reports to demonstrate the usefulness and the current limitations of the cell-based therapies.
Collapse
Affiliation(s)
- Yasuyuki Fujita
- Division of Vascular Regeneration, Unit of Regenerative Medicine, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation, Japan; Translational Research Informatics Center, Foundation for Biomedical Research and Innovation, Japan
| | - Atsuhiko Kawamoto
- Division of Vascular Regeneration, Unit of Regenerative Medicine, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation, Japan; Translational Research Informatics Center, Foundation for Biomedical Research and Innovation, Japan.
| |
Collapse
|
9
|
Ambasta RK, Kohli H, Kumar P. Multiple therapeutic effect of endothelial progenitor cell regulated by drugs in diabetes and diabetes related disorder. J Transl Med 2017; 15:185. [PMID: 28859673 PMCID: PMC5580204 DOI: 10.1186/s12967-017-1280-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/12/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Reduced levels of endothelial progenitor cells (EPCs) counts have been reported in diabetic mellitus (DM) patients and other diabetes-related disorder. EPCs are a circulating, bone marrow-derived cell population that appears to participate in vasculogenesis, angiogenesis and damage repair. These EPC may revert the damage caused in diabetic condition. We aim to identify several existing drugs and signaling molecule, which could alleviate or improve the diabetes condition via mobilizing and increasing EPC number as well as function. MAIN BODY Accumulated evidence suggests that dysregulation of EPC phenotype and function may be attributed to several signaling molecules and cytokines in DM patients. Hyperglycemia alone, through the overproduction of reactive oxygen species (ROS) via eNOS and NOX, can induce changes in gene expression and cellular behavior in diabetes. Furthermore, reports suggest that EPC telomere shortening via increased oxidative DNA damage may play an important role in the pathogenesis of coronary artery disease in diabetic patients. In this review, different type of EPC derived from different sources has been discussed along with cell-surface marker. The reduced number and immobilized EPC in diabetic condition have been mobilized for the therapeutic purpose via use of existing, and novel drugs have been discussed. Hence, evidence list of all types of drugs that have been reported to target the same pathway which affect EPC number and function in diabetes has been reviewed. Additionally, we highlight that proteins are critical in diabetes via polymorphism and inhibitor studies. Ultimately, a lucid pictorial explanation of diabetic and normal patient signaling pathways of the collected data have been presented in order to understand the complex signaling mystery underlying in the diseased and normal condition. CONCLUSION Finally, we conclude on eNOS-metformin-HSp90 signaling and its remedial effect for controlling the EPC to improve the diabetic condition for delaying diabetes-related complication. Altogether, the review gives a holistic overview about the elaborate therapeutic effect of EPC regulated by novel and existing drugs in diabetes and diabetes-related disorder.
Collapse
Affiliation(s)
- Rashmi K. Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, DTU, Delhi, India
| | - Harleen Kohli
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, DTU, Delhi, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, DTU, Delhi, India
| |
Collapse
|
10
|
Mistriotis P, Andreadis ST. Vascular aging: Molecular mechanisms and potential treatments for vascular rejuvenation. Ageing Res Rev 2017; 37:94-116. [PMID: 28579130 DOI: 10.1016/j.arr.2017.05.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 05/22/2017] [Accepted: 05/25/2017] [Indexed: 12/14/2022]
Abstract
Aging is the main risk factor contributing to vascular dysfunction and the progression of vascular diseases. In this review, we discuss the causes and mechanisms of vascular aging at the tissue and cellular level. We focus on Endothelial Cell (EC) and Smooth Muscle Cell (SMC) aging due to their critical role in mediating the defective vascular phenotype. We elaborate on two categories that contribute to cellular dysfunction: cell extrinsic and intrinsic factors. Extrinsic factors reflect systemic or environmental changes which alter EC and SMC homeostasis compromising vascular function. Intrinsic factors induce EC and SMC transformation resulting in cellular senescence. Replenishing or rejuvenating the aged/dysfunctional vascular cells is critical to the effective repair of the vasculature. As such, this review also elaborates on recent findings which indicate that stem cell and gene therapies may restore the impaired vascular cell function, reverse vascular aging, and prolong lifespan.
Collapse
Affiliation(s)
- Panagiotis Mistriotis
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
| | - Stelios T Andreadis
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA; Department of Biomedical Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA; Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14203, USA.
| |
Collapse
|
11
|
McNamee EN, Biette KA, Hammer J, Harris R, Miyazawa H, Lee JJ, Furuta GT, Masterson JC. Targeting granulocyte-macrophage colony-stimulating factor in epithelial and vascular remodeling in experimental eosinophilic esophagitis. Allergy 2017; 72:1232-1242. [PMID: 27926989 DOI: 10.1111/all.13105] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) is a chronic antigen-mediated clinicopathologic disease of the esophagus characterized by an eosinophil-predominant inflammatory infiltrate. A clinical hallmark is extensive tissue remodeling including basal zone hyperplasia, fibrosis, and angiogenesis. However, the cellular mechanisms responsible for these processes are not fully defined. We hypothesized that targeting granulocyte-macrophage colony-stimulating factor (GM-CSF; an agonist cytokine linked with eosinophil survival and activation) would be protective in a preclinical model of EoE. METHODS Eosinophilic esophagitis-like esophageal inflammation was induced in the L2-IL5OXA EoE mouse model, and GM-CSF production was assessed by mRNA and protein analyses. Granulocyte-macrophage colony-stimulating factor-receptor-alpha expression patterns were examined by flow cytometric and immunofluorescence analysis. L2-IL5OXA EoE mice were treated with anti-GM-CSF neutralizing antibody or isotype control and assessed for histopathological indices of eosinophilia, epithelial hyperplasia, and angiogenesis by immunohistochemistry and RT-PCR. RESULTS Significantly increased levels of esophageal GM-CSF expression was detected in the L2-IL5OXA mouse EoE model during active inflammation. Granulocyte-macrophage colony-stimulating factor-receptor-alpha was predominantly expressed on esophageal eosinophils during EoE, in addition to select cells within the lamina propria. Anti-GM-CSF neutralization in L2-IL5OXA EoE mice resulted in a significant diminution of epithelial eosinophilia in addition to basal cell hyperplasia and vascular remodeling. This treatment response was independent of effects on esophageal eosinophil maturation or activation. CONCLUSION Granulocyte-macrophage colony-stimulating factor is a potential therapeutic target to reduce esophageal eosinophilia and remodeling.
Collapse
Affiliation(s)
- E. N. McNamee
- Department of Pediatrics; Gastrointestinal Eosinophilic Diseases Program; Section of Pediatric Gastroenterology, Hepatology and Nutrition; University of Colorado School of Medicine; Aurora CO USA
- Digestive Health Institute; Children's Hospital Colorado; Aurora CO USA
- Department of Medicine; Mucosal Inflammation Program; University of Colorado School of Medicine; Aurora CO USA
- Department of Anesthesiology; University of Colorado School of Medicine; Aurora CO USA
| | - K. A. Biette
- Department of Pediatrics; Gastrointestinal Eosinophilic Diseases Program; Section of Pediatric Gastroenterology, Hepatology and Nutrition; University of Colorado School of Medicine; Aurora CO USA
- Digestive Health Institute; Children's Hospital Colorado; Aurora CO USA
- Department of Medicine; Mucosal Inflammation Program; University of Colorado School of Medicine; Aurora CO USA
| | - J. Hammer
- Department of Pediatrics; Gastrointestinal Eosinophilic Diseases Program; Section of Pediatric Gastroenterology, Hepatology and Nutrition; University of Colorado School of Medicine; Aurora CO USA
- Digestive Health Institute; Children's Hospital Colorado; Aurora CO USA
- Department of Medicine; Mucosal Inflammation Program; University of Colorado School of Medicine; Aurora CO USA
| | - R. Harris
- Department of Pediatrics; Gastrointestinal Eosinophilic Diseases Program; Section of Pediatric Gastroenterology, Hepatology and Nutrition; University of Colorado School of Medicine; Aurora CO USA
- Digestive Health Institute; Children's Hospital Colorado; Aurora CO USA
- Department of Medicine; Mucosal Inflammation Program; University of Colorado School of Medicine; Aurora CO USA
| | - H. Miyazawa
- Department of Pediatrics; Gastrointestinal Eosinophilic Diseases Program; Section of Pediatric Gastroenterology, Hepatology and Nutrition; University of Colorado School of Medicine; Aurora CO USA
- Digestive Health Institute; Children's Hospital Colorado; Aurora CO USA
| | - J. J. Lee
- Department of Biochemistry and Molecular Biology; Mayo Clinic; Scottsdale AZ USA
| | - G. T. Furuta
- Department of Pediatrics; Gastrointestinal Eosinophilic Diseases Program; Section of Pediatric Gastroenterology, Hepatology and Nutrition; University of Colorado School of Medicine; Aurora CO USA
- Digestive Health Institute; Children's Hospital Colorado; Aurora CO USA
- Department of Medicine; Mucosal Inflammation Program; University of Colorado School of Medicine; Aurora CO USA
| | - J. C. Masterson
- Department of Pediatrics; Gastrointestinal Eosinophilic Diseases Program; Section of Pediatric Gastroenterology, Hepatology and Nutrition; University of Colorado School of Medicine; Aurora CO USA
- Digestive Health Institute; Children's Hospital Colorado; Aurora CO USA
- Department of Medicine; Mucosal Inflammation Program; University of Colorado School of Medicine; Aurora CO USA
| |
Collapse
|
12
|
Effects of gamma-low dose irradiation on skin flap survival in rats. Phys Med 2017; 40:104-109. [DOI: 10.1016/j.ejmp.2017.07.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 07/16/2017] [Accepted: 07/22/2017] [Indexed: 11/17/2022] Open
|
13
|
Ollauri-Ibáñez C, López-Novoa JM, Pericacho M. Endoglin-based biological therapy in the treatment of angiogenesis-dependent pathologies. Expert Opin Biol Ther 2017; 17:1053-1063. [PMID: 28656781 DOI: 10.1080/14712598.2017.1346607] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Alterations in the process of angiogenesis, either by excess or by defect, are present in different common pathologies. For this reason, great efforts are being made toward the development of pro- and anti-angiogenic therapies. Since endoglin levels are enhanced in tissues undergoing angiogenesis, and changes in its expression lead to alterations in vessel formation, endoglin has become an ideal target for these types of therapies. Areas covered: In this review, the role of endoglin in angiogenesis is summarized. In addition, the authors review pro- and anti-angiogenic therapies that are currently being used and new approaches that target endoglin. The article includes therapies that are both in preclinical and clinical development. Expert opinion: Endoglin is a very good target for anti-angiogenic therapy, as demonstrated by the positive results obtained with anti-endoglin antibodies. However, although endoglin in pro-angiogenic therapies has been successful in vitro, its use has not yet reached clinical settings. Moreover, the authors believe that establishing the exact role of endoglin in angiogenesis is essential and that this should be the next step in this field in the coming years.
Collapse
Affiliation(s)
- Claudia Ollauri-Ibáñez
- a Department of Physiology and Pharmacology , University of Salamanca , Salamanca , Spain.,b Biomedical Research Institute of Salamanca (IBSAL) , Salamanca , Spain
| | - José M López-Novoa
- a Department of Physiology and Pharmacology , University of Salamanca , Salamanca , Spain.,b Biomedical Research Institute of Salamanca (IBSAL) , Salamanca , Spain
| | - Miguel Pericacho
- a Department of Physiology and Pharmacology , University of Salamanca , Salamanca , Spain.,b Biomedical Research Institute of Salamanca (IBSAL) , Salamanca , Spain
| |
Collapse
|
14
|
Shimizu Y, Sato S, Koyamatsu J, Yamanashi H, Nagayoshi M, Kadota K, Kawashiri SY, Maeda T. Association between high-density lipoprotein-cholesterol and hypertension in relation to circulating CD34-positive cell levels. J Physiol Anthropol 2017; 36:26. [PMID: 28619079 PMCID: PMC5472883 DOI: 10.1186/s40101-017-0143-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 06/07/2017] [Indexed: 12/22/2022] Open
Abstract
Background Although high-density lipoprotein-cholesterol (HDL) level is inversely correlated with cardiovascular events, HDL is also reported to be positively associated with hypertension, which is a known endothelial impairment factor. Since HDL mediates important protective actions on the vascular endothelium by increasing the number of circulating endothelial progenitor cells (CD34-positive cells), the level of circulating CD34-positive cells should influence the association between HDL and hypertension. Methods To investigate the association between HDL and hypertension in relation to the level of circulating CD34-positive cells, we conducted a cross-sectional study of 477 elderly men aged 60–69 years who participated in general health checkup. Results HDL was found to be significantly positively associated with hypertension in subjects with a high level of circulating CD34-positive cells, while no significant association was observed for subjects with low circulating CD34-positive cells. Known cardiovascular risk factors adjusted odds (ORs) and 95% confidence intervals (CIs) of hypertension for increments of one standard deviation (SD) in HDL (13.8 mg/dL) were 1.44 (1.06, 1.96) for subjects with a high level of circulating CD34-positive cells and 0.87 (0.63, 1.19) for subjects with low circulating CD34-positive cells. We also revealed a significant association between HDL level and CD34-positive cell level on hypertension, with fully adjusted p values for the effect of this interaction on hypertension at 0.022. Conclusions Independent of known cardiovascular risk factors, HDL was found to be positively associated with hypertension in subjects with a high level of circulating CD34-positive cells but not for subjects with low circulating CD34-positive cells.
Collapse
Affiliation(s)
- Yuji Shimizu
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan. .,Department of Cardiovascular Disease Prevention, Osaka Center for Cancer and Cardiovascular Disease Prevention, Osaka, Japan.
| | - Shimpei Sato
- Research and Clinical Center for Yusho and Dioxin, Kyusyu University, Fukuoka, Japan
| | - Jun Koyamatsu
- Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hirotomo Yamanashi
- Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Mako Nagayoshi
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Koichiro Kadota
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shin-Ya Kawashiri
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takahiro Maeda
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
15
|
Zigdon-Giladi H, Elimelech R, Michaeli-Geller G, Rudich U, Machtei EE. Safety profile and long-term engraftment of human CD31 + blood progenitors in bone tissue engineering. Cytotherapy 2017; 19:895-908. [PMID: 28495397 DOI: 10.1016/j.jcyt.2017.03.079] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/23/2017] [Accepted: 03/24/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) participate in angiogenesis and induce favorable micro-environments for tissue regeneration. The efficacy of EPCs in regenerative medicine is extensively studied; however, their safety profile remains unknown. Therefore, our aims were to evaluate the safety profile of human peripheral blood-derived EPCs (hEPCs) and to assess the long-term efficacy of hEPCs in bone tissue engineering. METHODS hEPCs were isolated from peripheral blood, cultured and characterized. β tricalcium phosphate scaffold (βTCP, control) or 106 hEPCs loaded onto βTCP were transplanted in a nude rat calvaria model. New bone formation and blood vessel density were analyzed using histomorphometry and micro-computed tomography (CT). Safety of hEPCs using karyotype analysis, tumorigenecity and biodistribution to target organs was evaluated. RESULTS On the cellular level, hEPCs retained their karyotype during cell expansion (seven passages). Five months following local hEPC transplantation, on the tissue and organ level, no inflammatory reaction or dysplastic change was evident at the transplanted site or in distant organs. Direct engraftment was evident as CD31 human antigens were detected lining vessel walls in the transplanted site. In distant organs human antigens were absent, negating biodistribution. Bone area fraction and bone height were doubled by hEPC transplantation without affecting mineral density and bone architecture. Additionally, local transplantation of hEPCs increased blood vessel density by nine-fold. CONCLUSIONS Local transplantation of hEPCs showed a positive safety profile. Furthermore, enhanced angiogenesis and osteogenesis without mineral density change was found. These results bring us one step closer to first-in-human trials using hEPCs for bone regeneration.
Collapse
Affiliation(s)
- Hadar Zigdon-Giladi
- Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus, Haifa, Israel; Research Institute for Bone Repair, Rambam Health Care Campus, Haifa, Israel; The Rappaport Family Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| | - Rina Elimelech
- Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus, Haifa, Israel; Research Institute for Bone Repair, Rambam Health Care Campus, Haifa, Israel
| | - Gal Michaeli-Geller
- Research Institute for Bone Repair, Rambam Health Care Campus, Haifa, Israel
| | - Utai Rudich
- Orthopedic Department, Rambam Health Care Campus, Haifa, Israel
| | - Eli E Machtei
- Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus, Haifa, Israel; Research Institute for Bone Repair, Rambam Health Care Campus, Haifa, Israel; The Rappaport Family Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
16
|
Hakami NY, Ranjan AK, Hardikar AA, Dusting GJ, Peshavariya HM. Role of NADPH Oxidase-4 in Human Endothelial Progenitor Cells. Front Physiol 2017; 8:150. [PMID: 28386230 PMCID: PMC5362645 DOI: 10.3389/fphys.2017.00150] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 02/24/2017] [Indexed: 01/21/2023] Open
Abstract
Introduction: Endothelial progenitor cells (EPCs) display a unique ability to promote angiogenesis and restore endothelial function in injured blood vessels. NADPH oxidase 4 (NOX4)-derived hydrogen peroxide (H2O2) serves as a signaling molecule and promotes endothelial cell proliferation and migration as well as protecting against cell death. However, the role of NOX4 in EPC function is not completely understood. Methods: EPCs were isolated from human saphenous vein and mammary artery discarded during bypass surgery. NOX4 gene and protein expression in EPCs were measured by real time-PCR and Western blot analysis respectively. NOX4 gene expression was inhibited using an adenoviral vector expressing human NOX4 shRNA (Ad-NOX4i). H2O2 production was measured by Amplex red assay. EPC migration was evaluated using a transwell migration assay. EPC proliferation and viability were measured using trypan blue counts. Results: Inhibition of NOX4 using Ad-NOX4i reduced Nox4 gene and protein expression as well as H2O2 formation in EPCs. Inhibition of NOX4-derived H2O2 decreased both proliferation and migration of EPCs. Interestingly, pro-inflammatory cytokine tumor necrosis factor alpha (TNFα) decreased NOX4 expression and reduced survival of EPCs. However, the survival of EPCs was further diminished by TNF-α in NOX4-knockdown cells, suggesting that NOX4 has a protective role in EPCs. Conclusion: These findings suggest that NOX4-type NADPH oxidase is important for proliferation and migration functions of EPCs and protects against pro-inflammatory cytokine induced EPC death. These properties of NOX4 may facilitate the efficient function of EPCs which is vital for successful neovascularization.
Collapse
Affiliation(s)
- Nora Y Hakami
- Centre for Eye Research Australia, Royal Victorian Eye and Ear HospitalEast Melbourne, VIC, Australia; Ophthalmology, University of Melbourne, Department of SurgeryEast Melbourne, VIC, Australia; Department of Pharmacology and Therapeutics, University of MelbourneMelbourne, VIC, Australia; Faculty of Applied Medical Sciences, King Abdulaziz UniversityJeddah, Saudi Arabia
| | - Amaresh K Ranjan
- Cardiology, Icahn School of Medicine at Mount Sinai Hospital New York, NY, USA
| | - Anandwardhan A Hardikar
- Diabetes and Islet Biology, NHMRC Clinical Trials Centre, University of Sydney Sydney, NSW, Australia
| | - Greg J Dusting
- Centre for Eye Research Australia, Royal Victorian Eye and Ear HospitalEast Melbourne, VIC, Australia; Ophthalmology, University of Melbourne, Department of SurgeryEast Melbourne, VIC, Australia
| | - Hitesh M Peshavariya
- Centre for Eye Research Australia, Royal Victorian Eye and Ear HospitalEast Melbourne, VIC, Australia; Ophthalmology, University of Melbourne, Department of SurgeryEast Melbourne, VIC, Australia
| |
Collapse
|
17
|
Vascular and perivascular niches, but not the osteoblastic niche, are numerically restored following allogeneic hematopoietic stem cell transplantation in patients with aplastic anemia. Int J Hematol 2017; 106:71-81. [PMID: 28303517 DOI: 10.1007/s12185-017-2217-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/09/2017] [Accepted: 03/09/2017] [Indexed: 12/19/2022]
Abstract
Bone marrow (BM) niches, including the osteoblastic, vascular, and perivascular niches, are numerically impaired in patients with aplastic anemia (AA). It remains unclear whether these niches are numerically restored in AA patients after allogenic hematopoietic stem cell transplantation (allo-HSCT). To investigate changes in BM niches, we monitored 52 patients with AA who had undergone allo-HSCT and performed immunohistochemical studies of BM niches using antibodies against CD34, CD146, and osteopontin. After allo-HSCT, patients with AA exhibited a remarkable increase in the number of cellular elements in the BM niches, including the vascular and perivascular cells. However, no significant differences in endosteal cells were detected. We explored the cause of this restoration by analyzing the origin of BM mesenchymal stem cells (BM-MSCs) and the expression of cytokines in BM plasma. STR-PCR revealed that the BM-MSCs were derived from the host, not the donor. In addition, significantly elevated levels of vascular endothelial growth factor (VEGF) were found after allo-HSCT. Our data indicates that vascular and perivascular niches are numerically restored, but the endosteal niche remains numerically impaired in patients with AA after allo-HSCT, and that levels of VEGF, but not donor-derived BM-MSCs, may correlate with the restoration of BM niches.
Collapse
|
18
|
Thej C, Ramadasse B, Walvekar A, Majumdar AS, Balasubramanian S. Development of a surrogate potency assay to determine the angiogenic activity of Stempeucel®, a pooled, ex-vivo expanded, allogeneic human bone marrow mesenchymal stromal cell product. Stem Cell Res Ther 2017; 8:47. [PMID: 28245882 PMCID: PMC5331748 DOI: 10.1186/s13287-017-0488-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 12/07/2016] [Accepted: 01/24/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) have emerged as a more beneficial alternative to conventional therapy and may offer a potential cure for unmet medical needs. MSCs are known to possess strong immunomodulatory and anti-inflammatory properties. Moreover, they promote angiogenesis and tissue regeneration through the secretion of trophic factors. For these reasons, the past decade witnessed a sharp increase in the number of clinical trials conducted with stem cells for various vascular diseases requiring angiogenesis. In this study, we evaluated the in vitro angiogenic potency of Stempeucel®, which is an allogeneic pooled human bone marrow-derived mesenchymal stromal cell (phBMMSC) product. We previously established the safety of Stempeucel® in our pre-clinical studies, and clinical trials conducted for critical limb ischaemia and acute myocardial infarction. METHODS Because the proposed mechanism of action of phBMMSCs is mainly through the secretion of pro-angiogenic cytokines, we developed a surrogate potency assay by screening various batches of large-scale expanded phBMMSCs for the expression of angiogenic factors and cytokines through gene expression and growth factor analyses, followed by in vitro functional assays. RESULTS The well characterized angiogenic vascular endothelial growth factor (VEGF) was selected and quantified in twenty six manufactured batches of phBMMSCs to establish consistency following the United States Food and Drug Administration recommendations. According to recommendations 21 CFR 211.165(e) and 211.194(a)(2), we also established and documented the specificity and reproducibility of the test methods employed through validation. Moreover, we also attempted to elucidate the mechanism of action of the cell population to ensure appropriate biological activity. The functional role of VEGF has been established through in vitro angiogenic assays and a dose-dependent correlation was observed with in vitro functional results. CONCLUSIONS The data generated from this study suggest the selection of VEGF as a single surrogate marker to test the angiogenic potency of phBMMSCs. Our study reports the quantification of VEGF in twenty six batches of large-scale manufactured phBMMSCs, and a concentration-dependent correlation of secreted VEGF to endothelial cell functions of migration, proliferation and tube formation, in the conditioned medium obtained from nine phBMMSC batches. To our cognizance, this is the first study in which a single angiogenic factor (VEGF) has been qualified as a surrogate potency marker through all three in vitro functional assays to determine the angiogenic potency of the phBMMSC population.
Collapse
Affiliation(s)
- Charan Thej
- Stempeutics Research Pvt. Ltd, Akshay Tech Park, #72 & 73, 2nd Floor, EPIP Zone, Phase 1, Bangalore, Whitefield, 560066, India.,Manipal University, Manipal, Karnataka, India
| | - Balamurugan Ramadasse
- Stempeutics Research Pvt. Ltd, Akshay Tech Park, #72 & 73, 2nd Floor, EPIP Zone, Phase 1, Bangalore, Whitefield, 560066, India
| | - Ankita Walvekar
- Stempeutics Research Pvt. Ltd, Akshay Tech Park, #72 & 73, 2nd Floor, EPIP Zone, Phase 1, Bangalore, Whitefield, 560066, India
| | - Anish S Majumdar
- Stempeutics Research Pvt. Ltd, Akshay Tech Park, #72 & 73, 2nd Floor, EPIP Zone, Phase 1, Bangalore, Whitefield, 560066, India.
| | - Sudha Balasubramanian
- Stempeutics Research Pvt. Ltd, Akshay Tech Park, #72 & 73, 2nd Floor, EPIP Zone, Phase 1, Bangalore, Whitefield, 560066, India.
| |
Collapse
|
19
|
Tamma R, Ribatti D. Bone Niches, Hematopoietic Stem Cells, and Vessel Formation. Int J Mol Sci 2017; 18:ijms18010151. [PMID: 28098778 PMCID: PMC5297784 DOI: 10.3390/ijms18010151] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/14/2016] [Accepted: 01/09/2017] [Indexed: 02/06/2023] Open
Abstract
Bone marrow (BM) is a source of hematopoietic stem cells (HSCs). HSCs are localized in both the endosteum, in the so-called endosteal niche, and close to thin-walled and fenestrated sinusoidal vessel in the center of BM, in the so-called vascular niche. HSCs give rise to all types of mature blood cells through a process finely controlled by numerous signals emerging from the bone marrow niches where HSCs reside. This review will focus on the description of the role of BM niches in the control of the fate of HSCs and will also highlight the role of the BM niches in the regulation of vasculogenesis and angiogenesis. Moreover, alterations of the signals in niche microenvironment are involved in many aspects of tumor progression and vascularization and further knowledge could provide the basis for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, 70124 Bari, Italy.
- National Cancer Institute Giovanni Paolo II, 70124 Bari, Italy.
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, 70124 Bari, Italy.
- National Cancer Institute Giovanni Paolo II, 70124 Bari, Italy.
| |
Collapse
|
20
|
Gebhard C, Rhéaume E, Berry C, Brand G, Kernaleguen AE, Théberge-Julien G, Alam MA, Lee CYW, Boileau L, Chabot-Blanchet M, Guertin MC, Lavoie MA, Grégoire J, Ibrahim R, L'Allier P, Tardif JC. Beneficial Effects of Reconstituted High-Density Lipoprotein (rHDL) on Circulating CD34+ Cells in Patients after an Acute Coronary Syndrome. PLoS One 2017; 12:e0168448. [PMID: 28060837 PMCID: PMC5218493 DOI: 10.1371/journal.pone.0168448] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 12/01/2016] [Indexed: 12/16/2022] Open
Abstract
Background High-density lipoproteins (HDL) favorably affect endothelial progenitor cells (EPC). Circulating progenitor cell level and function are impaired in patients with acute coronary syndrome (ACS). This study investigates the short-term effects of reconstituted HDL (rHDL) on circulating progenitor cells in patients with ACS. Methods and Findings The study population consisted of 33 patients with recent ACS: 20 patients from the ERASE trial (randomized to receive 4 weekly intravenous infusions of CSL-111 40 mg/kg or placebo) and 13 additional patients recruited as controls using the same enrolment criteria. Blood was collected from 16 rHDL (CSL-111)-treated patients and 17 controls at baseline and at 6–7 weeks (i.e. 2–3 weeks after the fourth infusion of CSL-111 in ERASE). CD34+ and CD34+/kinase insert domain receptor (KDR+) progenitor cell counts were analyzed by flow cytometry. We found preserved CD34+ cell counts in CSL-111-treated subjects at follow-up (change of 1.6%), while the number of CD34+ cells was reduced (-32.9%) in controls (p = 0.017 between groups). The level of circulating SDF-1 (stromal cell-derived factor-1), a chemokine involved in progenitor cell recruitment, increased significantly (change of 21.5%) in controls, while it remained unchanged in CSL-111-treated patients (p = 0.031 between groups). In vitro exposure to CSL-111 of early EPC isolated from healthy volunteers significantly increased CD34+ cells, reduced early EPC apoptosis and enhanced their migration capacity towards SDF-1. Conclusions The relative increase in circulating CD34+ cells and the low SDF-1 levels observed following rHDL infusions in ACS patients point towards a role of rHDL in cardiovascular repair mechanisms.
Collapse
Affiliation(s)
- Catherine Gebhard
- Montreal Heart Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Eric Rhéaume
- Montreal Heart Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Colin Berry
- Montreal Heart Institute, Montreal, Quebec, Canada
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | | | | | | | | | - Laurianne Boileau
- Montreal Heart Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | | | | | - Marc-André Lavoie
- Montreal Heart Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Jean Grégoire
- Montreal Heart Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Réda Ibrahim
- Montreal Heart Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Philippe L'Allier
- Montreal Heart Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Jean-Claude Tardif
- Montreal Heart Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
21
|
Novel protective effects of pulsed electromagnetic field ischemia/reperfusion injury rats. Biosci Rep 2016; 36:BSR20160082. [PMID: 27780890 PMCID: PMC5137536 DOI: 10.1042/bsr20160082] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 10/11/2016] [Accepted: 10/17/2016] [Indexed: 01/02/2023] Open
Abstract
Pulsed electromagnetic field (PEMF) treatment protected ischaemia/reperfusion (I/R) injury from apoptosis via B-cell lymphoma 2 (Bcl-2), Bax and nitric oxide (NO) releasing. Extracorporeal pulsed electromagnetic field (PEMF) has shown the ability to regenerate tissue by promoting cell proliferation. In the present study, we investigated for the first time whether PEMF treatment could improve the myocardial ischaemia/reperfusion (I/R) injury and uncovered its underlying mechanisms. In our study, we demonstrated for the first time that extracorporeal PEMF has a novel effect on myocardial I/R injury. The number and function of circulating endothelial progenitor cells (EPCs) were increased in PEMF treating rats. The in vivo results showed that per-treatment of PEMF could significantly improve the cardiac function in I/R injury group. In addition, PEMF treatment also reduced the apoptosis of myocardial cells by up-regulating the expression of anti-apoptosis protein B-cell lymphoma 2 (Bcl-2) and down-regulating the expression of pro-apoptosis protein (Bax). In vitro, the results showed that PEMF treatment could significantly reduce the apoptosis and reactive oxygen species (ROS) levels in primary neonatal rat cardiac ventricular myocytes (NRCMs) induced by hypoxia/reoxygenation (H/R). In particular, PEMF increased the phosphorylation of protein kinase B (Akt) and endothelial nitric oxide synthase (eNOS), which might be closely related to attenuated cell apoptosis by increasing the releasing of nitric oxide (NO). Therefore, our data indicated that PEMF could be a potential candidate for I/R injury.
Collapse
|
22
|
van Beijnum JR, Pieters W, Nowak-Sliwinska P, Griffioen AW. Insulin-like growth factor axis targeting in cancer and tumour angiogenesis - the missing link. Biol Rev Camb Philos Soc 2016; 92:1755-1768. [PMID: 27779364 DOI: 10.1111/brv.12306] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 09/15/2016] [Accepted: 09/21/2016] [Indexed: 12/14/2022]
Abstract
Numerous molecular players in the process of tumour angiogenesis have been shown to offer potential for therapeutic targeting. Initially denoted to be involved in malignant transformation and tumour progression, the insulin-like growth factor (IGF) signalling axis has been subject to therapeutic interference, albeit with limited clinical success. More recently, IGFs and their receptors have received attention for their contribution to tumour angiogenesis, which offers novel therapeutic opportunities. Here we review the contribution of this signalling axis to tumour angiogenesis, the mechanisms of resistance to therapy and the interplay with other pro-angiogenic pathways, to offer insight in the renewed interest in the application of IGF axis targeting agents in anti-cancer combination therapies.
Collapse
Affiliation(s)
- Judy R van Beijnum
- Department of Medical Oncology, Angiogenesis Laboratory, VU University Medical Center, PO box 7057, 1007 MB, Amsterdam, The Netherlands
| | - Wietske Pieters
- Department of Medical Oncology, Angiogenesis Laboratory, VU University Medical Center, PO box 7057, 1007 MB, Amsterdam, The Netherlands
| | - Patrycja Nowak-Sliwinska
- School of Pharmaceutical Sciences, University of Geneva (UNIGE), Rue Michel-Servet 1, 1211 Geneva 4, Switzerland
| | - Arjan W Griffioen
- Department of Medical Oncology, Angiogenesis Laboratory, VU University Medical Center, PO box 7057, 1007 MB, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Suzuki Y, Tada-Oikawa S, Hayashi Y, Izuoka K, Kataoka M, Ichikawa S, Wu W, Zong C, Ichihara G, Ichihara S. Single- and double-walled carbon nanotubes enhance atherosclerogenesis by promoting monocyte adhesion to endothelial cells and endothelial progenitor cell dysfunction. Part Fibre Toxicol 2016; 13:54. [PMID: 27737702 PMCID: PMC5064793 DOI: 10.1186/s12989-016-0166-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 10/05/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The use of carbon nanotubes has increased lately. However, the cardiovascular effect of exposure to carbon nanotubes remains elusive. The present study investigated the effects of pulmonary exposure to single-walled carbon nanotubes (SWCNTs) and double-walled carbon nanotubes (DWCNTs) on atherosclerogenesis using normal human aortic endothelial cells (HAECs) and apolipoprotein E-deficient (ApoE-/-) mice, a model of human atherosclerosis. METHODS HAECs were cultured and exposed to SWCNTs or DWCNTs for 16 h. ApoE-/- mice were exposed to SWCNTs or DWCNTs (10 or 40 μg/mouse) once every other week for 10 weeks by pharyngeal aspiration. RESULTS Exposure to CNTs increased the expression level of adhesion molecule (ICAM-1) and enhanced THP-1 monocyte adhesion to HAECs. ApoE-/- mice exposed to CNTs showed increased plaque area in the aorta by oil red O staining and up-regulation of ICAM-1 expression in the aorta, compared with vehicle-treated ApoE-/- mice. Endothelial progenitor cells (EPCs) are mobilized from the bone marrow into the circulation and subsequently migrate to the site of endothelial damage and repair. Exposure of ApoE-/- mice to high-dose SWCNTs or DWCNTs reduced the colony-forming units of EPCs in the bone marrow and diminished their migration function. CONCLUSION The results suggested that SWCNTs and DWCNTs enhanced atherosclerogenesis by promoting monocyte adhesion to endothelial cells and inducing EPC dysfunction.
Collapse
Affiliation(s)
- Yuka Suzuki
- Graduate School of Regional Innovation Studies, Mie University, 1577 Kurimamachiya-cho, Tsu, 514-8507, Japan
| | - Saeko Tada-Oikawa
- Graduate School of Regional Innovation Studies, Mie University, 1577 Kurimamachiya-cho, Tsu, 514-8507, Japan
| | - Yasuhiko Hayashi
- Graduate School of Natural Science and Technology, Okayama University, Okayama, Japan
| | - Kiyora Izuoka
- Graduate School of Regional Innovation Studies, Mie University, 1577 Kurimamachiya-cho, Tsu, 514-8507, Japan
| | - Misa Kataoka
- Graduate School of Regional Innovation Studies, Mie University, 1577 Kurimamachiya-cho, Tsu, 514-8507, Japan
| | - Shunsuke Ichikawa
- Graduate School of Regional Innovation Studies, Mie University, 1577 Kurimamachiya-cho, Tsu, 514-8507, Japan
| | - Wenting Wu
- Department of Occupational and Environmental Health, Nagoya Univeristy Graduate School of Medicine, Nagoya, Japan
| | - Cai Zong
- Department of Occupational and Environmental Health, Nagoya Univeristy Graduate School of Medicine, Nagoya, Japan
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Tokyo Univeristy of Science, Noda, Japan
| | - Sahoko Ichihara
- Graduate School of Regional Innovation Studies, Mie University, 1577 Kurimamachiya-cho, Tsu, 514-8507, Japan.
| |
Collapse
|
24
|
Van Pham P, Vu NB, Truong MTH, Huynh OT, Nguyen HT, Pham HL, Phan NK. Hepatocyte growth factor improves direct reprogramming of fibroblasts towards endothelial progenitor cells via ETV2 transduction. BIOMEDICAL RESEARCH AND THERAPY 2016. [DOI: 10.7603/s40730-016-0045-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
25
|
Kalathil SG, Lugade AA, Iyer R, Miller A, Thanavala Y. Endothelial progenitor cell number and ERK phosphorylation serve as predictive and prognostic biomarkers in advanced hepatocellular carcinoma patients treated with sorafenib. Oncoimmunology 2016; 5:e1226718. [PMID: 27853648 DOI: 10.1080/2162402x.2016.1226718] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 08/11/2016] [Accepted: 08/16/2016] [Indexed: 12/13/2022] Open
Abstract
Sorafenib is an oral anti-angiogenic multi-kinase inhibitor used for systemic therapy in patients with advanced hepatocellular carcinoma (HCC) who are not suitable candidates for surgery or liver transplantation. An earlier study conducted with HCC tumor tissue suggested that ERK phosphorylation (pERK), a downstream target of sorafenib, may serve as a potential biomarker for therapeutic efficacy of sorafenib. However, no study thus far has utilized a minimal invasive procedure to predict HCC patient responsiveness to sorafenib. We evaluated the biomarker utility of circulating endothelial progenitor cells (EPCs) frequency and intracellular pERK levels in EPCs in peripheral blood obtained pre- and post-sorafenib therapy or after transarterial chemoembolistaion (TACE). A statistically significant reduction in the level of ERK phosphorylation and in the absolute number of EPCs was detected following in vivo sorafenib treatment (p < 0 .01 for both). In contrast, the decrease in the level of ERK phosphorylation and EPC number was either marginally significant or insignificant in patients treated with TACE (p = 0.05 and 0.06, respectively). In vitro sorafenib treatment of pre- and post-samples from the same patient cohort inhibited ERK phosphorylation levels in EPCs and decreased the number of EPCs at all doses tested (p = 0.01). Our findings support that the evaluation of both the circulating EPC frequency and the level of ERK phosphorylation in EPCs may serve as potential non-invasive biomarkers of sorafenib efficacy, both as predictor of treatment outcome and efficacy during drug treatment.
Collapse
Affiliation(s)
| | - Amit Anand Lugade
- Department of Immunology, Roswell Park Cancer Institute , Buffalo, NY, USA
| | - Renuka Iyer
- Department of Medicine, Roswell Park Cancer Institute , Buffalo, NY, USA
| | - Austin Miller
- Department of Biostatistics & Bioinformatics, Roswell Park Cancer Institute , Buffalo, NY, USA
| | - Yasmin Thanavala
- Department of Immunology, Roswell Park Cancer Institute , Buffalo, NY, USA
| |
Collapse
|
26
|
Jazwa A, Florczyk U, Grochot-Przeczek A, Krist B, Loboda A, Jozkowicz A, Dulak J. Limb ischemia and vessel regeneration: Is there a role for VEGF? Vascul Pharmacol 2016; 86:18-30. [PMID: 27620809 DOI: 10.1016/j.vph.2016.09.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 07/24/2016] [Accepted: 09/07/2016] [Indexed: 11/18/2022]
Abstract
Vascular endothelial growth factor (VEGF), as an endothelial cell-specific mitogen, is crucial for new blood vessels formation. Atherosclerosis affecting the cardiovascular system causes ischemia and functio laesa in tissues supplied by the occluded vessels. When such a situation occurs in the lower extremities, it causes critical limb ischemia (CLI) often requiring leg amputation. Low oxygen tension leads to upregulation of hypoxia-regulated genes (i.e. VEGF), that should help to restore the impaired blood flow. In CLI these rescue mechanisms are, however, often inefficient. Moreover, there are many contradictory reports showing either induction, no changes or even down-regulation of VEGF in specimens taken from patients with CLI, as well as in samples collected from animals subjected to hindlimb ischemia. Additionally, taking into account numerous experimental and clinical data demonstrating rather insufficient therapeutic potential of VEGF, we called into question the role of this protein in limb ischemia and vessel regeneration. In this review we are also summarizing several aspects which can influence VEGF expression and its measurement in the ischemic tissues.
Collapse
Affiliation(s)
- Agnieszka Jazwa
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| | - Urszula Florczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Bart Krist
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Agnieszka Loboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland; Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
27
|
Chhokar V, Tucker AL. Angiogenesis: Basic Mechanisms and Clinical Applications. Semin Cardiothorac Vasc Anesth 2016. [DOI: 10.1177/108925320300700304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The development and maintenance of an adequate vascular supply is critical for the viability of normal and neoplastic tissues. Angiogenesis, the development of new blood vessels from preexisting capillary networks, plays an important role in a number of physiologic and pathologic processes, including reproduction, wound repair, inflammatory diseases, and tumor growth. Angiogenesis involves sequential steps that are triggered in response to angiogenic growth factors released by inflammatory, mesenchymal, or tumor cells that act as ligands for endothelial cell receptor tyrosine kinases. Stimulated endothelial cells detach from neighboring cells and migrate, proliferate, and form tubes. The immature tubes are subsequently invested and stabilized by pericytes or smooth muscle cells. Angiogenesis depends upon complex interactions among various classes of molecules, including adhesion molecules, proteases, structural proteins, cell surface receptors, and growth factors. The therapeutic manipulation of angiogenesis targeted against ischemic and neoplastic diseases has been investigated in preclinical animal models and in clinical trials. Proangiogenic trials that have stimulated vessel growth in ischemic coronary or peripheral tissues through expression, delivery, or stimulated release of growth factors have shown efficacy in animal models and mixed results in human clinical trials. Antiangiogenic trials have used strategies to block the function of molecules critical for new vessel growth or maturation in the treatment of a variety of malignancies, mostly with results less encouraging than those seen in preclinical models. Pro-and antiangiogenic clinical trials demonstrate that strategies for optimal drug delivery, dosing schedules, patient selection, and endpoint measurements need further investigation and refinement before the therapeutic manipulation of angiogenesis will realize its full clinical potential.
Collapse
Affiliation(s)
- Vikram Chhokar
- Department of Internal Medicine, Salem VA Health System, Roanoke, Virginia
| | - Amy L. Tucker
- Department of Internal Medicine, Cardiovascular Division; Cardiovascular Research Center; Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
28
|
Endothelial progenitor cells accelerate the resolution of deep vein thrombosis. Vascul Pharmacol 2016; 83:10-6. [DOI: 10.1016/j.vph.2015.07.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 06/27/2015] [Accepted: 07/11/2015] [Indexed: 11/23/2022]
|
29
|
Chatzikyrkou C, Bahlmann FH, Sushakova N, Scurt FG, Menne J, Nawroth P, Mertens PR, Haller H. Low-dose erythropoietin promotes wound-healing of ulcers in diabetics: Evidence from a phase-IIa clinical study. DIABETES & METABOLISM 2016; 42:466-470. [PMID: 27324461 DOI: 10.1016/j.diabet.2016.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 05/23/2016] [Accepted: 05/24/2016] [Indexed: 11/26/2022]
Affiliation(s)
- C Chatzikyrkou
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany; Department of Nephrology, Hypertension, Diabetology and Endocrinology, University Hospital of Magdeburg, Leipziger Street 44, 39112 Magdeburg, Germany.
| | - F H Bahlmann
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - N Sushakova
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - F G Scurt
- Department of Nephrology, Hypertension, Diabetology and Endocrinology, University Hospital of Magdeburg, Leipziger Street 44, 39112 Magdeburg, Germany
| | - J Menne
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - P Nawroth
- Department of Internal Medicine I and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | - P R Mertens
- Department of Nephrology, Hypertension, Diabetology and Endocrinology, University Hospital of Magdeburg, Leipziger Street 44, 39112 Magdeburg, Germany
| | - H Haller
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| |
Collapse
|
30
|
Witzigmann D, Quagliata L, Schenk SH, Quintavalle C, Terracciano LM, Huwyler J. Variable asialoglycoprotein receptor 1 expression in liver disease: Implications for therapeutic intervention. Hepatol Res 2016; 46:686-96. [PMID: 26422581 DOI: 10.1111/hepr.12599] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/07/2015] [Accepted: 09/19/2015] [Indexed: 02/08/2023]
Abstract
AIM One of the most promising strategies for the treatment of liver diseases is targeted drug delivery via the asialoglycoprotein receptor (ASGPR). The success of this approach heavily depends on the ASGPR expression level on parenchymal liver cells. In this study, we assessed the mRNA and protein expression levels of the major receptor subunit, ASGR1, in hepatocytes both in vitro and in vivo. METHODS In vitro, various liver cancer-derived cell lines were evaluated. In vivo, we screened the ASGR1 mRNA on 59 hepatocellular carcinoma and matched non-neoplastic tissue using RNA microarray. In addition, 350 human liver specimens of patients with hepatocellular carcinoma or non-neoplastic liver diseases were screened for ASGR1 protein level using tissue microarray analysis. RESULTS Our data reveal that the ASGR1 mRNA expression directly correlates with the protein level. We demonstrate that the ASGR1 expression is upregulated in cirrhotic specimens and is significantly decreased with increasing hepatocellular carcinoma grade. CONCLUSION Because the ASGR1 expression levels are variable between patients, our findings suggest that ASGPR-based targeting strategies should be combined with ASGPR-companion diagnostics to maximize clinical benefit.
Collapse
Affiliation(s)
- Dominik Witzigmann
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Luca Quagliata
- Institute of Pathology, Molecular Pathology Division, University Hospital of Basel, Basel, Switzerland
| | - Susanne H Schenk
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Cristina Quintavalle
- Institute of Pathology, Molecular Pathology Division, University Hospital of Basel, Basel, Switzerland
| | - Luigi M Terracciano
- Institute of Pathology, Molecular Pathology Division, University Hospital of Basel, Basel, Switzerland
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| |
Collapse
|
31
|
Chang TT, Wu TC, Huang PH, Chen JS, Lin LY, Lin SJ, Chen JW. Aliskiren directly improves endothelial progenitor cell function from Type II diabetic patients. Eur J Clin Invest 2016; 46:544-54. [PMID: 27062013 DOI: 10.1111/eci.12632] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 04/08/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Endothelial progenitor cell (EPC) functions are impaired in the presence of diabetes mellitus. Aliskiren is a direct renin inhibitor, which is expected to modify proangiogenic cells. This study aimed to investigate whether and how aliskiren could improve the function of EPCs from patients with type II diabetes (T2DM). MATERIALS AND METHODS Endothelial progenitor cells fibronectin adhesion assay, chamber assay and in vitro tube formation assay were used to estimate the degree of EPC adhesion, migration and tube formation abilities. EPC protein and mRNA expressions were evaluated by Western blot and quantitative RT-PCR, respectively. EPC vascular endothelial growth factor (VEGF) and (pro)renin receptor ((P)RR) expression was knocked down by VEGF and (P)RR siRNA. RESULTS Aliskiren (0·1 or 10 μM) dose-dependently improved functions and increased both VEGF and stromal cell-derived factor-1α (SDF-1α) expression of EPCs from patients with T2DM or EPCs from healthy volunteers and treated with high glucose. Transfection with VEGF siRNA significantly reduced the aliskiren-induced SDF-1α expression. Furthermore, (P)RR siRNA transfection impaired the aliskiren-induced VEGF and SDF-1 expression. CONCLUSIONS The results show that aliskiren improved EPC function from patients with T2DM in a dose-dependent manner probably via the (P)RR and VEGF/SDF-1α-related mechanisms.
Collapse
Affiliation(s)
- Ting-Ting Chang
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Tao-Cheng Wu
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Po-Hsun Huang
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Jia-Shiong Chen
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan.,Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Liang-Yu Lin
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan.,Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shing-Jong Lin
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jaw-Wen Chen
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan.,Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
32
|
Suen CM, Zhai A, Lalu MM, Welsh C, Levac BM, Fergusson D, McIntyre L, Stewart DJ. Efficacy and safety of regenerative cell therapy for pulmonary arterial hypertension in animal models: a preclinical systematic review protocol. Syst Rev 2016; 5:89. [PMID: 27225668 PMCID: PMC4880876 DOI: 10.1186/s13643-016-0265-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 05/10/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a rare disease (15 cases per million) that is characterized by widespread loss of the pulmonary microcirculation and elevated pulmonary vascular resistance leading to pathological right ventricular remodeling and ultimately right heart failure. Regenerative cell therapies (i.e., therapies involving cells with stem or progenitor-like properties) could potentially restore the effective lung microcirculation and provide a curative therapy for PAH. Preclinical evidence suggests that regenerative cell therapy using endothelial progenitor cells or mesenchymal stem cells may be beneficial in the treatment of PAH. These findings have led to the completion of a small number of human clinical trials, albeit with modest effect compared to animal studies. The objective of this systematic review is to compare the efficacy and safety of regenerative cell therapies in preclinical models of PAH as well as assess study quality to inform future clinical studies. METHODS We will include preclinical studies of PAH in which a regenerative cell type was administered and outcomes compared to a disease control. The primary outcome will be pulmonary hemodynamics as assessed by measurement of right ventricular systolic pressure and/or mean pulmonary arterial pressure. Secondary outcomes will include mortality, survival, right ventricular remodeling, pulmonary vascular resistance, cardiac output, cardiac index, pulmonary acceleration time, tricuspid annular systolic excursion, and right ventricular wall thickness. Electronic searches of MEDLINE and EMBASE databases will be constructed and reviewed by the Peer Review of Electronic Search Strategies (PRESS) process. Search results will be screened independently in duplicate. Data from eligible studies will be extracted, pooled, and analyzed using random effects models. Risk of bias will be assessed using the SYstematic Review Centre for Laboratory animal Experimentation (SYRCLE) risk of bias tool, and individual study reporting will be assessed according to an itemized checklist based on the Animal Research: Reporting of In vivo Experiments (ARRIVE) guidelines. DISCUSSION This systematic review will examine the efficacy and safety of regenerative cell therapy in preclinical models of PAH. As well, the literature will be assessed for study quality and risk of bias. The results will guide the design of future clinical trials and preclinical animal studies. SYSTEMATIC REVIEW REGISTRATION CAMARADES ( http://www.dcn.ed.ac.uk/camarades/SyRF/Protocols.htm ).
Collapse
Affiliation(s)
- Colin M Suen
- Regenerative Medicine Program, The Ottawa Hospital Research Institute, 501 Smyth Rd, Ottawa, ON, K1H8L6, Canada.,Department of Cell and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Alex Zhai
- Regenerative Medicine Program, The Ottawa Hospital Research Institute, 501 Smyth Rd, Ottawa, ON, K1H8L6, Canada
| | - Manoj M Lalu
- Regenerative Medicine Program, The Ottawa Hospital Research Institute, 501 Smyth Rd, Ottawa, ON, K1H8L6, Canada.,Department of Anesthesiology, The Ottawa Hospital, The Ottawa Hospital Research Institute, Ottawa, Canada
| | | | - Brendan M Levac
- Department of Medicine, University of Ottawa, Ottawa, Canada
| | - Dean Fergusson
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Medicine, University of Ottawa, Ottawa, Canada.,Department of Surgery, University of Ottawa, Ottawa, Canada.,Department of Epidemiology and Community Medicine, University of Ottawa, Ottawa, Canada
| | - Lauralyn McIntyre
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Medicine, University of Ottawa, Ottawa, Canada
| | - Duncan J Stewart
- Regenerative Medicine Program, The Ottawa Hospital Research Institute, 501 Smyth Rd, Ottawa, ON, K1H8L6, Canada. .,Department of Cell and Molecular Medicine, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
33
|
Divergent modulation of normal and neoplastic stem cells by thrombospondin-1 and CD47 signaling. Int J Biochem Cell Biol 2016; 81:184-194. [PMID: 27163531 DOI: 10.1016/j.biocel.2016.05.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 04/27/2016] [Accepted: 05/04/2016] [Indexed: 01/19/2023]
Abstract
Thrombospondin-1 is a secreted matricellular protein that regulates the differentiation and function of many cell types. Thrombospondin-1 is not required for embryonic development, but studies using lineage-committed adult stem cells have identified positive and negative effects of thrombospondin-1 on stem cell differentiation and self-renewal and identified several thrombospondin-1 receptors that mediate these responses. Genetic studies in mice reveal a broad inhibitory role of thrombospondin-1 mediated by its receptor CD47. Cells and tissues lacking thrombospondin-1 or CD47 exhibit an increased capacity for self-renewal associated with increased expression of the stem cell transcription factors c-Myc, Sox2, Klf4, and Oct4. Thrombospondin-1 inhibits expression of these transcription factors in a CD47-dependent manner. However, this regulation differs in some neoplastic cells. Tumor initiating/cancer stem cells express high levels of CD47, but in contrast to nontransformed stem cells CD47 signaling supports cancer stem cells. Suppression of CD47 expression in cancer stem cells or ligation of CD47 by function blocking antibodies or thrombospondin-1 results in loss of self-renewal. Therefore, the therapeutic CD47 antagonists that are in clinical development for stimulating innate anti-tumor immunity may also inhibit tumor growth by suppressing cancer stem cells. These and other therapeutic modulators of thrombospondin-1 and CD47 signaling may also have applications in regenerative medicine to enhance the function of normal stem cells.
Collapse
|
34
|
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
|
35
|
Takizawa S, Nagata E, Nakayama T, Masuda H, Asahara T. Recent Progress in Endothelial Progenitor Cell Culture Systems: Potential for Stroke Therapy. Neurol Med Chir (Tokyo) 2016; 56:302-9. [PMID: 27041632 PMCID: PMC4908073 DOI: 10.2176/nmc.ra.2016-0027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Endothelial progenitor cells (EPCs) participate in endothelial repair and angiogenesis due to their abilities to differentiate into endothelial cells and to secrete protective cytokines and growth factors. Consequently, there is considerable interest in cell therapy with EPCs isolated from peripheral blood to treat various ischemic injuries. Quality and quantity-controlled culture systems to obtain mononuclear cells enriched in EPCs with well-defined angiogenic and anti-inflammatory phenotypes have recently been developed, and increasing evidence from animal models and clinical trials supports the idea that transplantation of EPCs contributes to the regenerative process in ischemic organs and is effective for the therapy of ischemic cerebral injury. Here, we briefly describe the general characteristics of EPCs, and we review recent developments in culture systems and applications of EPCs and EPC-enriched cell populations to treat ischemic stroke.
Collapse
Affiliation(s)
- Shunya Takizawa
- Department of Neurology, Tokai University School of Medicine
| | | | | | | | | |
Collapse
|
36
|
Guillevic O, Ferratge S, Pascaud J, Driancourt C, Boyer-Di-Ponio J, Uzan G. A Novel Molecular and Functional Stemness Signature Assessing Human Cord Blood-Derived Endothelial Progenitor Cell Immaturity. PLoS One 2016; 11:e0152993. [PMID: 27043207 PMCID: PMC4820260 DOI: 10.1371/journal.pone.0152993] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 03/22/2016] [Indexed: 01/20/2023] Open
Abstract
Endothelial Colony Forming Cells (ECFCs), a distinct population of Endothelial Progenitor Cells (EPCs) progeny, display phenotypic and functional characteristics of endothelial cells while retaining features of stem/progenitor cells. Cord blood-derived ECFCs (CB-ECFCs) have a high clonogenic and proliferative potentials and they can acquire different endothelial phenotypes, this requiring some plasticity. These properties provide angiogenic and vascular repair capabilities to CB-ECFCs for ischemic cell therapies. However, the degree of immaturity retained by EPCs is still confused and poorly defined. Consequently, to better characterize CB-ECFC stemness, we quantified their clonogenic potential and demonstrated that they were reprogrammed into induced pluripotent stem cells (iPSCs) more efficiently and rapidly than adult endothelial cells. Moreover, we analyzed the transcriptional profile of a broad gene panel known to be related to stem cells. We showed that, unlike mature endothelial cells, CB-ECFCs expressed genes involved in the maintenance of embryonic stem cell properties such as DNMT3B, GDF3 or SOX2. Thus, these results provide further evidence and tools to appreciate EPC-derived cell stemness. Moreover this novel stem cell transcriptional signature of ECFCs could help better characterizing and ranging EPCs according to their immaturity profile.
Collapse
Affiliation(s)
| | | | | | | | | | - Georges Uzan
- INSERM U972, hôpital Paul Brousse, Villejuif, France
- * E-mail:
| |
Collapse
|
37
|
Circulating endothelial progenitor cells may predict outcomes in adult patients with severe sepsis in the emergency department. Clin Chim Acta 2016; 455:1-6. [DOI: 10.1016/j.cca.2016.01.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 01/05/2016] [Accepted: 01/16/2016] [Indexed: 12/18/2022]
|
38
|
Saboo A, Rathnayake A, Vangaveti VN, Malabu UH. Wound healing effects of dipeptidyl peptidase-4 inhibitors: An emerging concept in management of diabetic foot ulcer-A review. Diabetes Metab Syndr 2016; 10:113-119. [PMID: 25990796 DOI: 10.1016/j.dsx.2015.04.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Dipeptidyl peptidase-4 (DPP-4) inhibitors have a well-known effect on glycaemic control in patients with diabetes but little is known on their wound healing role in this group of population. This paper reviews the effects of DPP-4 inhibitors on wound healing of diabetic foot ulcers. METHODS Published data on effects and mechanism of DDP-4 inhibitors on wound healing were derived from Medline, PubMed and Google Scholar search of English language literature from 1994 to 2014 using the key words such as "DPP-4 inhibitors", "endothelial healing" "diabetes" and "chronic ulcers". RESULTS DPP-4 inhibitors show a potential benefit in processes of wound healing in diabetic chronic foot ulcers. The enzyme inhibitors promote recruitment of endothelial progenitor cells and allow the final scaffolding of wounds. Furthermore DPP-4 inhibitors augment angiogenesis and have widespread effects on optimising the immune response to persistent hypoxia in chronic diabetes wounds. CONCLUSION DPP-4 inhibitors show promise in the local wound healing of diabetic foot ulcers in addition to its already established glycaemic control. In the light of high rate of amputations due to non-healing ulcers with profound psychological and economical liability, more investigations on the usefulness of DPP-4 inhibitors in the high risk diabetes population are needed.
Collapse
Affiliation(s)
- Apoorva Saboo
- Translational Research in Endocrinology and Diabetes [TREAD], College of Medicine and Dentistry, James Cook University, The Townsville Hospital, 100 Angus Smith Drive, Douglas 4814, QLD, Australia
| | - Ayeshmanthe Rathnayake
- Translational Research in Endocrinology and Diabetes [TREAD], College of Medicine and Dentistry, James Cook University, The Townsville Hospital, 100 Angus Smith Drive, Douglas 4814, QLD, Australia
| | - Venkat N Vangaveti
- Translational Research in Endocrinology and Diabetes [TREAD], College of Medicine and Dentistry, James Cook University, The Townsville Hospital, 100 Angus Smith Drive, Douglas 4814, QLD, Australia
| | - Usman H Malabu
- Translational Research in Endocrinology and Diabetes [TREAD], College of Medicine and Dentistry, James Cook University, The Townsville Hospital, 100 Angus Smith Drive, Douglas 4814, QLD, Australia.
| |
Collapse
|
39
|
De Angelis A, Urbanek K, Cappetta D, Piegari E, Ciuffreda LP, Rivellino A, Russo R, Esposito G, Rossi F, Berrino L. Doxorubicin cardiotoxicity and target cells: a broader perspective. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2016; 2:2. [PMID: 33530140 PMCID: PMC7837148 DOI: 10.1186/s40959-016-0012-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 02/18/2016] [Indexed: 12/12/2022]
Abstract
The cardiotoxicity of doxorubicin is becoming an interdisciplinary point of interest given a growing population of cancer survivors. The complex and not completely understood pathogenesis of this complication makes difficult to design successful preventive or curative measures. Although cardiomyocyte has been considered a classical cellular target, other cells including various types of undifferentiated cells are involved in myocardial homeostasis. Such perspective may shed light on previously unrecognized aspects of cardiotoxicity and promote new experimental and clinical cardioprotective strategies. In this review, different cellular targets of doxorubicin are discussed with the focus on cardiac progenitor cells, oxidative stress, DNA damage, senescence and apoptosis all of which contribute to their compromised functional properties.
Collapse
Affiliation(s)
- Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Konrad Urbanek
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Donato Cappetta
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Elena Piegari
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Loreta Pia Ciuffreda
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Alessia Rivellino
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Rosa Russo
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Grazia Esposito
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| |
Collapse
|
40
|
Um J, Jung N, Chin S, Cho Y, Choi S, Park KS. Substance P enhances EPC mobilization for accelerated wound healing. Wound Repair Regen 2016; 24:402-10. [PMID: 26749197 DOI: 10.1111/wrr.12403] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 12/28/2015] [Indexed: 12/21/2022]
Abstract
Wound healing is essential for the survival and tissue homeostasis of unicellular and multicellular organisms. The current study demonstrated that the neuropeptide substance P (SP) accelerated the wound healing process, particularly in the skin. Subcutaneous treatment of SP accelerated wound closing, increased the population of α-smooth muscle actin positive myofibroblasts, and increased extracellular matrix deposition at the wound site. Moreover, SP treatment enhances angiogenesis without a local increase in the expression levels of vascular endothelial growth factor and stromal cell-derived factor-1. Importantly, SP treatment increased both the population of circulating endothelial progenitor cells in the peripheral blood and in CD31 positive cells in Matrigel plugs. The tube forming potential of endothelial cells was also enhanced by SP treatment. The results suggested that the subcutaneous injection of SP accelerated the wound healing in the skin via better reconstitution of blood vessels, which possibly followed an increase in the systemic mobilization of endothelial progenitor cells and a more effective assembly of endothelial cells into tubes.
Collapse
Affiliation(s)
- Jihyun Um
- Graduate School of Biotechnology, Kyung Hee University, Yongin-si, South Korea
| | - Nunggum Jung
- Graduate School of Biotechnology, Kyung Hee University, Yongin-si, South Korea
| | - Sukbum Chin
- Graduate School of Biotechnology, Kyung Hee University, Yongin-si, South Korea
| | - Younggil Cho
- Department of Genetic Engineering, Kyung Hee University, Yongin-si, South Korea
| | - Sanghyuk Choi
- Department of Genetic Engineering, Kyung Hee University, Yongin-si, South Korea
| | - Ki-Sook Park
- East-West Medical Research Institute/College of Medicine, Kyung Hee University, Seoul, Yongin-si, 02447, South Korea
| |
Collapse
|
41
|
Jang HK, Kim BS, Han J, Yoon JK, Lee JR, Jeong GJ, Shin JY. Therapeutic angiogenesis using tumor cell-conditioned medium. Biotechnol Prog 2016; 32:456-64. [DOI: 10.1002/btpr.2226] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 12/31/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Hyeon-Ki Jang
- Interdisciplinary Program for Bioengineering; Seoul National University; Seoul 151-744 Republic of Korea
| | - Byung-Soo Kim
- Interdisciplinary Program for Bioengineering; Seoul National University; Seoul 151-744 Republic of Korea
- School of Chemical and Biological Engineering; Seoul National University; Seoul 151-744 Republic of Korea
- Bio-MAX Inst.; Inst. for Chemical Processes, Seoul National University; Seoul 151-744 Republic of Korea
| | - Jin Han
- School of Chemical and Biological Engineering; Seoul National University; Seoul 151-744 Republic of Korea
| | - Jeong-Kee Yoon
- School of Chemical and Biological Engineering; Seoul National University; Seoul 151-744 Republic of Korea
| | - Ju-Ro Lee
- School of Chemical and Biological Engineering; Seoul National University; Seoul 151-744 Republic of Korea
| | - Gun-Jae Jeong
- School of Chemical and Biological Engineering; Seoul National University; Seoul 151-744 Republic of Korea
| | - Jung-Youn Shin
- School of Chemical and Biological Engineering; Seoul National University; Seoul 151-744 Republic of Korea
| |
Collapse
|
42
|
Klar AS, Güven S, Zimoch J, Zapiórkowska NA, Biedermann T, Böttcher-Haberzeth S, Meuli-Simmen C, Martin I, Scherberich A, Reichmann E, Meuli M. Characterization of vasculogenic potential of human adipose-derived endothelial cells in a three-dimensional vascularized skin substitute. Pediatr Surg Int 2016; 32:17-27. [PMID: 26621500 DOI: 10.1007/s00383-015-3808-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/09/2015] [Indexed: 12/24/2022]
Abstract
PURPOSE The need for clinically applicable skin substitutes continues to be a matter of fact. Hypothetically, a laboratory grown autologous skin analog with near normal architecture might be a suitable approach to yield both satisfactory functional and cosmetic long-term results. In this study, we explored the use of human endothelial cells derived from freshly isolated adipose stromal vascular fraction (SVF) in a three-dimensional (3D) co-culture model of vascularized bio-engineered skin substitute. METHODS The SVF was isolated from human white adipose tissue samples and keratinocytes from human skin biopsies. The SVF, in particular endothelial cells, were characterized using flow cytometry and immuofluorescence analysis. Endothelial and mesenchymal progenitors from the SVF formed blood capillaries after seeding into a 3D collagen type I hydrogel in vitro. Subsequently, human keratinocytes were seeded on the top of those hydrogels to develop a vascularized dermo-epidermal skin substitute. RESULTS Flow cytometric analysis of surface markers of the freshly isolated SVF showed the expression of endothelial markers (CD31, CD34, CD146), mesenchymal/stromal cell-associated markers (CD44, CD73, CD90, CD105), stem cell markers (CD49f, CD117, CD133), and additionally hematopoietic markers (CD14, CD15, CD45). Further analysis of white adipose-derived endothelial cells (watECs) revealed the co-expression of CD31, CD34, CD90, CD105, and partially CD146 on these cells. WatECs were separated from adipose-stromal cells (watASCs) using FACS sorting. WatASCs and watECs cultured separately in a 3D hydrogel for 3 weeks did not form any vascular structures. Only if co-cultured, both cell types aligned to develop a ramified vascular network in vitro with continuous endothelial lumen formation. Transplantation of those 3D-hydrogels onto immuno-incompetent rats resulted in a rapid connection of human capillaries with the host vessels and formation of functional, blood-perfused mosaic human-rat vessels within only 3-4 days. CONCLUSIONS Adipose tissue represents an attractive cell source due to the ease of isolation and abundance of endothelial as well as mesenchymal cell lineages. Adipose-derived SVF cells exhibit the ability to form microvascular structures in vitro and support the accelerated blood perfusion in skin substitutes in vivo when transplanted.
Collapse
Affiliation(s)
- Agnes S Klar
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Sinan Güven
- Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Jakub Zimoch
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Natalia A Zapiórkowska
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Sophie Böttcher-Haberzeth
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- Department of Surgery, University Children's Hospital Zurich, Steinwiesstrasse 75, 8032, Zurich, Switzerland
| | - Claudia Meuli-Simmen
- Department of Plastic, Reconstructive, Esthetical and Hand Surgery, Kantonsspital Aarau, Aarau, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Arnaud Scherberich
- Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Ernst Reichmann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Martin Meuli
- Department of Surgery, University Children's Hospital Zurich, Steinwiesstrasse 75, 8032, Zurich, Switzerland.
| |
Collapse
|
43
|
Kalyva A, Marketou ME, Parthenakis FI, Pontikoglou C, Kontaraki JE, Maragkoudakis S, Petousis S, Chlouverakis G, Papadaki HA, Vardas PE. Endothelial progenitor cells as markers of severity in hypertrophic cardiomyopathy. Eur J Heart Fail 2015; 18:179-84. [PMID: 26696595 DOI: 10.1002/ejhf.436] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 10/05/2015] [Accepted: 10/06/2015] [Indexed: 01/05/2023] Open
Abstract
AIMS Endothelial progenitor cells (EPCs) are bone marrow-derived cells that are mobilized into the circulation to migrate and differentiate into mature endothelial cells contributing to post-natal physiological and pathological neovascularization. In this study, we evaluated circulating EPCs in patients with hypertrophic cardiomyopathy (HCM) and examined a potential association with clinical parameters of the disease. METHODS AND RESULTS We included 40 HCM patients and 23 healthy individuals. Using flow cytometry we measured EPCs in peripheral blood as two subpopulations of CD45-/CD34+/VEGFR2+ and CD45-/CD34+/CD133+ cells. Circulating CD45-/CD34+/VEGFR2+ cells were significantly increased in HCM patients in comparison with the controls (0.000238 ± 0.0003136 vs. 0.000057 ± 0.0001316, respectively, P = 0.002). However, there was no significant difference in the number of circulating CD45-/CD34+/CD133+ cells (0.003079 ± 0.0033288 vs. 0.002065 ± 0.0022173, respectively, P = 0.153). The CD45-/CD34+/VEGFR2+ subpopulation revealed a moderate correlation with LV mass index (r = 0.35, P = 0.026), while both EPC subpopulation levels showed strong positive correlations with th E/e' ratio (r = 0.423, P = 0.007 for CD45-/CD34+/VEGFR2+ and r = 0.572, P < 0.001 for CD45-/CD34+/CD133+). CONCLUSION HCM patients showed an increased mobilization of EPCs compared with healthy individuals that correlated with diastolic dysfunction. Our findings may open up new dimensions in the pathophysiology, prognostication, and treatment of HCM.
Collapse
Affiliation(s)
- Athanasia Kalyva
- Molecular Cardiology Laboratory, School of Medicine, University of Crete, Greece
| | - Maria E Marketou
- Department of Cardiology, Heraklion University Hospital, Crete, Greece
| | | | | | - Joanna E Kontaraki
- Molecular Cardiology Laboratory, School of Medicine, University of Crete, Greece
| | | | | | | | - Helen A Papadaki
- Department of Haematology, Heraklion University Hospital, Crete, Greece
| | - Panos E Vardas
- Department of Cardiology, Heraklion University Hospital, Crete, Greece
| |
Collapse
|
44
|
Elbuken G, Tanriverdi F, Karaca Z, Eser B, Hasdiraz L, Unluhizarci K, Gokoglu A, Cetin A, Selcuklu A, Kelestimur F. Evaluation of peripheral blood CD34+ cell count in the acute phase of traumatic brain injury and chest trauma. Brain Inj 2015; 30:179-83. [PMID: 26649467 DOI: 10.3109/02699052.2015.1090015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIM To determine the impact of traumatic brain injury (TBI) and chest trauma (CT) on the number of peripheral blood (PB) stem cells in affected patients in comparison to normal controls. Additionally, the aim was to determine the relationship between CD34+ cell counts and TBI-induced hypothalamus-pituitary-adrenal axis dysfunction in the acute phase of trauma. PATIENTS AND METHOD Thirty patients with TBI, 12 patients with CT and 53 healthy subjects were included in the study. RESULTS CD34+ cell counts within the first 24-48 hours of TBI were found to be lower than those obtained on the 7(th) day of TBI and those in the healthy controls. CD34+ cell counts obtained on the 2(nd) day of CT were lower than those in the healthy group, but did not differ from those measured on the 7(th) day of CT. There was no correlation between CD34+ cell counts and serum total cortisol (STC) levels on the 2(nd) and 7(th) days in the TBI or CT groups. CONCLUSION An increase in CD34+ cell counts as observed on the 7(th) day in both TBI and CT groups suggested that CD34 changes were not specific to TBI. Moreover, this study showed for the first time that CD34 response was not affected by changes in cortisol levels induced by TBI and severity of TBI.
Collapse
Affiliation(s)
- Gulsah Elbuken
- a Department of Endocrinology and Metabolism , Erciyes University Medical School , Kayseri , Turkey
| | - Fatih Tanriverdi
- a Department of Endocrinology and Metabolism , Erciyes University Medical School , Kayseri , Turkey
| | - Zuleyha Karaca
- a Department of Endocrinology and Metabolism , Erciyes University Medical School , Kayseri , Turkey
| | - Bulent Eser
- b Department of Hematology , Erciyes University Medical School , Kayseri , Turkey
| | - Leyla Hasdiraz
- c Department of Chest Surgery , Erciyes University Medical School , Kayseri , Turkey
| | - Kursad Unluhizarci
- a Department of Endocrinology and Metabolism , Erciyes University Medical School , Kayseri , Turkey
| | - Abdulkerim Gokoglu
- d Department of Neurosurgery , Erciyes University Medical School , Kayseri , Turkey
| | - Aysun Cetin
- e Department of Biochemistry , Erciyes University Medical School , Kayseri , Turkey
| | - Ahmet Selcuklu
- d Department of Neurosurgery , Erciyes University Medical School , Kayseri , Turkey
| | - Fahrettin Kelestimur
- a Department of Endocrinology and Metabolism , Erciyes University Medical School , Kayseri , Turkey
| |
Collapse
|
45
|
Aso Y, Jojima T, Iijima T, Suzuki K, Terasawa T, Fukushima M, Momobayashi A, Hara K, Takebayashi K, Kasai K, Inukai T. Sitagliptin, a dipeptidyl peptidase-4 inhibitor, increases the number of circulating CD34⁺CXCR4⁺ cells in patients with type 2 diabetes. Endocrine 2015. [PMID: 26209038 DOI: 10.1007/s12020-015-0688-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We investigated the effects of sitagliptin, a dipeptidyl peptidase (DPP)-4 inhibitor, on the number of circulating CD34(+)CXCR4(+)cells, a candidate for endothelial progenitor cells (EPCs), plasma levels of stromal cell-derived factor (SDF)-1α, a ligand for CXCR4 receptor and a substrate for DPP-4, and plasma levels of interferon-inducible protein (IP)-10, for a substrate for DPP-4, in patients with type 2 diabetes. We studied 30 consecutive patients with type 2 diabetes who had poor glycemic control despite treatment with metformin and/or sulfonylurea. Thirty diabetic patients were randomized in a 2:1 ratio into a sitagliptin (50 mg/day) treatment group or an active placebo group (glimepiride 1 mg/day) for 12 weeks. Both groups showed similar improvements in glycemic control. The number of circulating CD34(+)CXCR4(+) cells was increased from 30.5 (20.0, 47.0)/10(6) cells at baseline to 55.5 (31.5, 80.5)/10(6) cells at 12 weeks of treatment with 50 mg/day sitagliptin (P = 0.0014), while showing no significant changes in patients treated with glimepiride. Plasma levels of SDF-1α and IP-10, both physiological substrates of endogenous DPP-4 and chemokines, were significantly decreased at 12 weeks of sitagliptin treatment. In conclusion, treatment with sitagliptin increased the number of circulating CD34(+)CXCR4(+) cells by approximately 2-fold in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Yoshimasa Aso
- Department of Endocrinology and Metabolism, Dokkyo Medical University, 880 Kita-Kobayashi, Shimotsuga, Mibu, Tochigi, 321-0293, Japan.
| | - T Jojima
- Department of Endocrinology and Metabolism, Dokkyo Medical University, 880 Kita-Kobayashi, Shimotsuga, Mibu, Tochigi, 321-0293, Japan
| | - T Iijima
- Department of Endocrinology and Metabolism, Dokkyo Medical University, 880 Kita-Kobayashi, Shimotsuga, Mibu, Tochigi, 321-0293, Japan
| | - K Suzuki
- Department of Endocrinology and Metabolism, Dokkyo Medical University, 880 Kita-Kobayashi, Shimotsuga, Mibu, Tochigi, 321-0293, Japan
| | - T Terasawa
- Department of Internal Medicine, Koshigaya Hospital, Dokkyo Medical University, Koshigaya, Saitama, Japan
| | - M Fukushima
- LSI Medience Corp., Itabashi-Ku, Tokyo, Japan
| | | | - K Hara
- Department of Internal Medicine, Koshigaya Hospital, Dokkyo Medical University, Koshigaya, Saitama, Japan
| | - K Takebayashi
- Department of Internal Medicine, Koshigaya Hospital, Dokkyo Medical University, Koshigaya, Saitama, Japan
| | - K Kasai
- Department of Medicine, Ishibashi General Hospital, Shimotsuke, Tochigi, Japan
| | - T Inukai
- Department of Internal Medicine, Koshigaya Hospital, Dokkyo Medical University, Koshigaya, Saitama, Japan
| |
Collapse
|
46
|
Balaji S, Han N, Moles C, Shaaban AF, Bollyky PL, Crombleholme TM, Keswani SG. Angiopoietin-1 improves endothelial progenitor cell-dependent neovascularization in diabetic wounds. Surgery 2015; 158:846-56. [PMID: 26266763 DOI: 10.1016/j.surg.2015.06.034] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Revised: 06/22/2015] [Accepted: 06/27/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND The diabetic phenotype of wound healing is in part characterized by impaired neovascularization and deficient endothelial progenitor cell (EPC) recruitment. Angiopoietin-1 (Ang-1) is a potent mobilizer of EPCs from the bone marrow (BM). A suggested mechanism for EPC mobilization from the BM is mediated by matrix metalloproteinase 9 (MMP-9) and stem cell factor (SCF). Taken together, we hypothesized that overexpression of Ang-1 in diabetic wounds will recruit EPCs and improve neovascularization and wound healing. METHODS An endothelial lineage BM-labeled murine model of diabetes was developed to track BM-derived EPCs. FVBN mice were lethally irradiated and then reconstituted with BM from syngeneic Tie2/LacZ donor mice. Diabetes was induced with streptozotocin. Dorsal wounds in BM-transplanted mice were treated with Ad-Ang-1, Ad-GFP, or phosphate-buffered saline. At day 7 after injury, wounds were harvested and analyzed. A similar experiment was conducted in EPC mobilization deficient MMP-9 -/- mice to determine whether the effects of Ang-1 were EPC-dependent. RESULTS Overexpression of Ang-1 resulted in greatly improved re-epithelialization, neovascularization, and EPC recruitment in diabetic BM-transplanted wounds at day 7. Ang-1 treatment resulted in increased serum levels of proMMP-9 and SCF but had no effect on vascular endothelial growth factor levels. According to our FACS results, peripheral blood EPC (CD34(+)/Cd133(+)/Flk1(+)) counts at day 3 after wounding showed impaired EPC mobilization in MMP-9 -/- mice compared with those of wild-type controls. EPC mobilization was rescued by SCF administration, validating this model for EPC-mobilization-deficient mechanistic studies. In MMP-9 -/- mice, Ad-Ang-1 accelerated re-epithelialization in a similar manner, but had no effect on neovascularization. CONCLUSION Our results show that Ang-1 administration results in improved neovascularization which is dependent on EPC recruitment and has direct effects on wound re-epithelialization. These data may represent a novel strategy to correct the phenotype of impaired diabetic neovascularization and may improve diabetic wound healing.
Collapse
Affiliation(s)
- Swathi Balaji
- Laboratory for Regenerative Wound Healing, Division of Pediatric, General, Thoracic and Fetal Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Nate Han
- Laboratory for Regenerative Wound Healing, Division of Pediatric, General, Thoracic and Fetal Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Chad Moles
- Laboratory for Regenerative Wound Healing, Division of Pediatric, General, Thoracic and Fetal Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Aimen F Shaaban
- Laboratory for Regenerative Wound Healing, Division of Pediatric, General, Thoracic and Fetal Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Paul L Bollyky
- Division of Infectious Diseases, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Timothy M Crombleholme
- Laboratory for Regenerative Wound Healing, Division of Pediatric, General, Thoracic and Fetal Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Center for Children's Surgery, Children's Hospital Colorado and the University of Colorado School of Medicine, Aurora, CO
| | - Sundeep G Keswani
- Laboratory for Regenerative Wound Healing, Division of Pediatric, General, Thoracic and Fetal Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.
| |
Collapse
|
47
|
Zhang J, Garrison JC, Poluektova LY, Bronich TK, Osna NA. Liver-targeted antiviral peptide nanocomplexes as potential anti-HCV therapeutics. Biomaterials 2015; 70:37-47. [PMID: 26298393 PMCID: PMC4562313 DOI: 10.1016/j.biomaterials.2015.08.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 08/05/2015] [Accepted: 08/07/2015] [Indexed: 02/05/2023]
Abstract
Great success in HCV therapy was achieved by the development of direct-acting antivirals (DAA). However, the unsolved issues such as high cost and genotype dependency drive us to pursue additional therapeutic agents to be used instead or in combination with DAA. The cationic peptide p41 is one of such candidates displaying submicromolar anti-HCV potency. By electrostatic coupling of p41 with anionic poly(amino acid)-based block copolymers, antiviral peptide nanocomplexes (APN) platform was developed to improve peptide stability and to reduce cytotoxicity associated with positive charge. Herein, we developed a facile method to prepare galactosylated Gal-APN and tested their feasibility as liver-specific delivery system. In vitro, Gal-APN displayed specific internalization in hepatoma cell lines. Even though liver-targeted and non-targeted APN displayed comparable antiviral activity, Gal-APN offered prominent advantages to prevent HCV association with lipid droplets and suppress intracellular expression of HCV proteins. Moreover, in vivo preferential liver accumulation of Gal-APN was revealed in the biodistribution study. Altogether, this work illustrates the potential of Gal-APN as a novel liver-targeted therapy against HCV.
Collapse
Affiliation(s)
- Jinjin Zhang
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jered C Garrison
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Larisa Y Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Tatiana K Bronich
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Natalia A Osna
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA; Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System Medical Center, 4101 Woolworth Avenue, Omaha, NE 68105, USA.
| |
Collapse
|
48
|
Joseph P, Ishai A, MacNabb M, Abdelbaky A, Lavender ZR, Ruskin J, Nahrendorf M, Tawakol A. Atrial fibrillation is associated with hematopoietic tissue activation and arterial inflammation. Int J Cardiovasc Imaging 2015; 32:113-9. [PMID: 26411879 DOI: 10.1007/s10554-015-0766-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 09/09/2015] [Indexed: 01/01/2023]
Abstract
Inflammation is associated with the development of atrial fibrillation (AF). Activity in hematopoietic tissues, which produce inflammatory leukocytes, is closely related to systemic inflammation, arterial inflammation and cardiovascular events, but its relationship to AF is unknown. Using 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) imaging, we examined the relationships between AF, splenic metabolic activity and vascular inflammation. We conducted a cross sectional study of 70 participants: 35 with AF, who were matched (by age, sex and history of active cancer) to 35 controls without AF. Splenic metabolic activity and vascular aortic inflammation were measured by the mean FDG maximum standard uptake values (SUV Max) by PET. We examined (1) the association between AF and splenic activity, and (2) AF and aortic inflammation. The mean age of the population was 68.13 (standard deviation (SD) 8.98) years and 46 (65 %) participants were male. Splenic activity was higher in AF participants [2.31 (SD 0.45) vs. 2.07 (SD 0.37), p = 0.024], and remained significant after adjusting for demographic and clinical covariates. Aortic inflammation was also higher in AF participants [2.22 (SD 0.44) vs. 1.91 (SD 0.44), p = 0.004], and remained significant on multivariable analysis. Aortic inflammation and splenic activity were highly correlated (Pearson R = 0.61, p < 0.001). Atrial fibrillation is associated with higher hematopoietic tissue activation and arterial inflammation. Further studies are needed to clarify the mechanisms by which this cardio-splenic axis is implicated in AF.
Collapse
Affiliation(s)
- Philip Joseph
- Cardiac MR PET CT Program, Division of Cardiac Imaging, Massachusetts General Hospital and Harvard Medical School, Suite 400-165 Cambridge St, Boston, MA, 02114, USA
- McMaster University, Hamilton, ON, Canada
| | - Amorina Ishai
- Cardiac MR PET CT Program, Division of Cardiac Imaging, Massachusetts General Hospital and Harvard Medical School, Suite 400-165 Cambridge St, Boston, MA, 02114, USA
| | - Megan MacNabb
- Cardiac MR PET CT Program, Division of Cardiac Imaging, Massachusetts General Hospital and Harvard Medical School, Suite 400-165 Cambridge St, Boston, MA, 02114, USA
| | - Amr Abdelbaky
- Cardiac MR PET CT Program, Division of Cardiac Imaging, Massachusetts General Hospital and Harvard Medical School, Suite 400-165 Cambridge St, Boston, MA, 02114, USA
| | - Zachary R Lavender
- Cardiac MR PET CT Program, Division of Cardiac Imaging, Massachusetts General Hospital and Harvard Medical School, Suite 400-165 Cambridge St, Boston, MA, 02114, USA
| | - Jeremy Ruskin
- Cardiac MR PET CT Program, Division of Cardiac Imaging, Massachusetts General Hospital and Harvard Medical School, Suite 400-165 Cambridge St, Boston, MA, 02114, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ahmed Tawakol
- Cardiac MR PET CT Program, Division of Cardiac Imaging, Massachusetts General Hospital and Harvard Medical School, Suite 400-165 Cambridge St, Boston, MA, 02114, USA.
| |
Collapse
|
49
|
Cardiac Nerve Growth Factor Overexpression Induces Bone Marrow-derived Progenitor Cells Mobilization and Homing to the Infarcted Heart. Mol Ther 2015; 23:1854-66. [PMID: 26354341 PMCID: PMC4700109 DOI: 10.1038/mt.2015.167] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 08/12/2015] [Indexed: 12/16/2022] Open
Abstract
Reparative response by bone marrow (BM)-derived progenitor cells (PCs) to ischemia is a multistep process that comprises the detachment from the BM endosteal niche through activation of osteoclasts and proteolytic enzymes (such as matrix metalloproteinases (MMPs)), mobilization to the circulation, and homing to the injured tissue. We previously showed that intramyocardial nerve growth factor gene transfer (NGF-GT) promotes cardiac repair following myocardial infarction (MI) in mice. Here, we investigate the impact of cardiac NGF-GT on postinfarction BM-derived PCs mobilization and homing at different time points after adenovirus-mediated NGF-GT in mice. Immunohistochemistry and flow cytometry newly illustrate the temporal profile of osteoclast and activation of MMP9, PCs expansion in the BM, and liberation/homing to the injured myocardium. NGF-GT amplified these responses and increased the BM levels of active osteoclasts and MMP9, which were not observed in MMP9-deficient mice. Taken together, our results suggest a novel role for NGF in BM-derived PCs mobilization/homing following MI.
Collapse
|
50
|
Li WD, Hu N, Lei FR, Wei S, Rong JJ, Zhuang H, Li XQ. Autophagy inhibits endothelial progenitor cells migration via the regulation of MMP2, MMP9 and uPA under normoxia condition. Biochem Biophys Res Commun 2015; 466:376-80. [PMID: 26363453 DOI: 10.1016/j.bbrc.2015.09.031] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 09/05/2015] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The aim of this study was to explore the role of autophagy on the regulation of endothelial progenitor cells (EPCs) migration under normoxic condition. METHODS After EPCs were isolated and characterized in vitro, we employed Atg5 knocking down and rapamycin to monitor the autophagy, and performed wound healing and transwell assay to assess the cell migration. On the mechanism, the expression of matrix metalloproteinases (MMPs) and urokinase type plasminogen activator (uPA) was evaluated. RESULTS Atg5 knocking down and rapamycin could respectively inhibit and enhance autophagy, which could result in significantly increased and decreased cell migration in wound healing and transwell assay under normoxic condition. Moreover, Atg5 knocking down could significantly increase the expression of MMP2, MMP9 and uPA in EPCs while rapamycin could decrease the expression of uPA and MMP9. In addition, the mTOR-P70 S6K pathway was also involved in EPCs migration regulation. CONCLUSIONS These results demonstrated that autophagy could regulate the EPCs migration through mTOR-P70 S6K pathway, and MMP2, MMP9 and uPA may also involve in the regulation mechanism.
Collapse
Affiliation(s)
- Wen-Dong Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Nan Hu
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Feng-Rui Lei
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Sen Wei
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian-Jie Rong
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hao Zhuang
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiao-Qiang Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|