1
|
Gudde AN, van Velthoven MJJ, Kouwer PHJ, Roovers JPWR, Guler Z. Injectable polyisocyanide hydrogel as healing supplement for connective tissue regeneration in an abdominal wound model. Biomaterials 2023; 302:122337. [PMID: 37793268 DOI: 10.1016/j.biomaterials.2023.122337] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/07/2023] [Accepted: 09/23/2023] [Indexed: 10/06/2023]
Abstract
In pelvic organ prolapse (POP) patients, the uterus, bladder and/or rectum descends into vagina due to weakened support tissues. High recurrence rates after POP surgery suggest an urgent need for improved surgical outcomes. Our aim is to promote connective tissue healing that results in stimulated tissue support functions by surgically applying a hydrogel functionalized with biological cues. We used known vaginal wound healing promoting factors (basic fibroblast growth factor, β-estradiol, adipose-derived stem cells) in the biomimetic and injectable polyisocyanide (PIC) hydrogel, which in itself induces regenerative vaginal fibroblast behavior. The regenerative capacity of injected PIC hydrogel, and the additional pro-regenerative effects of these bioactive factors was evaluated in abdominal wounds in rabbits. Assessment of connective tissue healing (tensile testing, histology, immunohistochemistry) revealed that injection with all PIC formulations resulted in a statistically significant stiffness and collagen increase over time, in contrast to sham. Histological evaluation indicated new tissue growth with moderate to mild immune activity at the hydrogel - tissue interface. The results suggest that PIC injection in an abdominal wound improves healing towards regaining load-bearing capacity, which encourages us to investigate application of the hydrogel in a more translational vaginal model for POP surgery in sheep.
Collapse
Affiliation(s)
- Aksel N Gudde
- Department of Obstetrics and Gynecology, Amsterdam University Medical Center-location AMC, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Amsterdam Reproduction and Development, Amsterdam University Medical Center-location AMC, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Melissa J J van Velthoven
- Department of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, the Netherlands; Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, the Netherlands
| | - Paul H J Kouwer
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, the Netherlands
| | - Jan-Paul W R Roovers
- Department of Obstetrics and Gynecology, Amsterdam University Medical Center-location AMC, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Amsterdam Reproduction and Development, Amsterdam University Medical Center-location AMC, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Zeliha Guler
- Department of Obstetrics and Gynecology, Amsterdam University Medical Center-location AMC, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Amsterdam Reproduction and Development, Amsterdam University Medical Center-location AMC, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands.
| |
Collapse
|
2
|
Kanaji Y, Ozcan I, Toya T, Gulati R, Young M, Kakuta T, Lerman LO, Lerman A. Circulating Progenitor Cells Are Associated With Bioprosthetic Aortic Valve Deterioration: A Preliminary Study. J Am Heart Assoc 2023; 12:e027364. [PMID: 36645093 PMCID: PMC9939063 DOI: 10.1161/jaha.122.027364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Background Mechanisms underlying bioprosthetic valve deterioration are multifactorial and incompletely elucidated. Reparative circulating progenitor cells, and conversely calcification-associated osteocalcin expressing circulating progenitor cells, have been linked to native aortic valve deterioration. However, their role in bioprosthetic valve deterioration remains elusive. This study sought to evaluate the contribution of different subpopulations of circulating progenitor cells in bioprosthetic valve deterioration. Methods and Results This single-center prospective study enrolled 121 patients who had peripheral blood mononuclear cells isolated before bioprosthetic aortic valve replacement and had an echocardiographic follow-up ≥2 years after the procedure. Using flow cytometry, fresh peripheral blood mononuclear cells were analyzed for the surface markers CD34, CD133, and osteocalcin. Bioprosthetic valve deterioration was evaluated by hemodynamic valve deterioration (HVD) using echocardiography, which was defined as an elevated mean transprosthetic gradient ≥30 mm Hg or at least moderate intraprosthetic regurgitation. Sixteen patients (13.2%) developed HVD during follow-up for a median of 5.9 years. Patients with HVD showed significantly lower levels of reparative CD34+CD133+ cells and higher levels of osteocalcin-positive cells than those without HVD (CD34+CD133+ cells: 125 [80, 210] versus 270 [130, 420], P=0.002; osteocalcin-positive cells: 3060 [523, 5528] versus 670 [180, 1930], P=0.005 respectively). Decreased level of CD34+CD133+ cells was a significant predictor of HVD (hazard ratio, 0.995 [95% CI, 0.990%-0.999%]). Conclusions Circulating levels of CD34+CD133+ cells and osteocalcin-positive cells were significantly associated with the subsequent occurrence of HVD in patients undergoing bioprosthetic aortic valve replacement. Circulating progenitor cells might play a vital role in the mechanism, risk stratification, and a potential therapeutic target for patients with bioprosthetic valve deterioration.
Collapse
Affiliation(s)
- Yoshihisa Kanaji
- Department of Cardiovascular MedicineRochesterMN,Division of Cardiovascular MedicineTsuchiura Kyodo General HospitalIbarakiJapan
| | - Ilke Ozcan
- Department of Cardiovascular MedicineRochesterMN
| | - Takumi Toya
- Department of Cardiovascular MedicineRochesterMN,Division of CardiologyNational Defense Medical CollegeTokorozawaJapan
| | - Rajiv Gulati
- Department of Cardiovascular MedicineRochesterMN
| | | | - Tsunekazu Kakuta
- Division of Cardiovascular MedicineTsuchiura Kyodo General HospitalIbarakiJapan
| | - Lilach O. Lerman
- Division of Nephrology and Hypertension, Mayo ClinicMayo ClinicRochesterMN
| | - Amir Lerman
- Department of Cardiovascular MedicineRochesterMN
| |
Collapse
|
3
|
Marshall AJ, Gaubert A, Kapoor A, Tan A, McIntosh E, Jang JY, Yew B, Ho JK, Blanken AE, Dutt S, Sible IJ, Li Y, Rodgers K, Nation DA. Blood-Derived Progenitor Cells Are Depleted in Older Adults with Cognitive Impairment: A Role for Vascular Resilience? J Alzheimers Dis 2023; 93:1041-1050. [PMID: 37154177 PMCID: PMC10258882 DOI: 10.3233/jad-220269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND Depletion of blood-derived progenitor cells, including so called "early endothelial progenitor cells", has been observed in individuals with early stage Alzheimer's disease relative to matched older control subjects. These findings could implicate the loss of angiogenic support from hematopoietic progenitors or endothelial progenitors in cognitive dysfunction. OBJECTIVE To investigate links between progenitor cell proliferation and mild levels of cognitive dysfunction. METHODS We conducted in vitro studies of blood-derived progenitor cells using blood samples from sixty-five older adults who were free of stroke or dementia. Peripheral blood mononuclear cells from venous blood samples were cultured in CFU-Hill media and the number of colony forming units were counted after 5 days in vitro. Neuropsychological testing was administered to all participants. RESULTS Fewer colony forming units were observed in samples from older adults with a Clinical Dementia Rating global score of 0.5 versus 0. Older adults whose samples developed fewer colony forming units exhibited worse performance on neuropsychological measures of memory, executive functioning, and language ability. CONCLUSION These data suggest blood progenitors may represent a vascular resilience marker related to cognitive dysfunction in older adults.
Collapse
Affiliation(s)
- Anisa J. Marshall
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Aimee Gaubert
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Arunima Kapoor
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Alick Tan
- Department of Clinical Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Elissa McIntosh
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Jung Yun Jang
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Belinda Yew
- Department of Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jean K. Ho
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Anna E. Blanken
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Shubir Dutt
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Isabel J. Sible
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Yanrong Li
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Kathleen Rodgers
- Center for Innovations in Brain Science, Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Daniel A. Nation
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
4
|
The Long Telling Story of "Endothelial Progenitor Cells": Where Are We at Now? Cells 2022; 12:cells12010112. [PMID: 36611906 PMCID: PMC9819021 DOI: 10.3390/cells12010112] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
Endothelial progenitor cells (EPCs): The name embodies years of research and clinical expectations, but where are we now? Do these cells really represent the El Dorado of regenerative medicine? Here, past and recent literature about this eclectic, still unknown and therefore fascinating cell population will be discussed. This review will take the reader through a temporal journey that, from the first discovery, will pass through years of research devoted to attempts at their definition and understanding their biology in health and disease, ending with the most recent evidence about their pathobiological role in cardiovascular disease and their recent applications in regenerative medicine.
Collapse
|
5
|
Quader S, Tanabe S, Cabral H. Abnormal Glycosylation in Cancer Cells and Cancer Stem Cells as a Therapeutic Target. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1393:141-156. [PMID: 36587306 DOI: 10.1007/978-3-031-12974-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Tumor resistance and recurrence have been associated with the presence of cancer stem cells (CSCs) in tumors. The functions and survival of the CSCs have been associated with several intracellular and extracellular features. Particularly, the abnormal glycosylation of these signaling pathways and markers of CSCs have been correlated with maintaining survival, self-renewal and extravasation properties. Here, we highlight the importance of glycosylation in promoting the stemness character of CSCs and the current strategies for targeting abnormal glycosylation toward generating effective therapies against the CSC population.
Collapse
Affiliation(s)
- Sabina Quader
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Shihori Tanabe
- Division of Risk Assessment, Center for Biological Safety and Research, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, 210-9501, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan.
| |
Collapse
|
6
|
Muggeridge D, Dodd J, Ross MD. CD34 + progenitors are predictive of mortality and are associated with physical activity in cardiovascular disease patients. Atherosclerosis 2021; 333:108-115. [PMID: 34340831 DOI: 10.1016/j.atherosclerosis.2021.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 06/17/2021] [Accepted: 07/08/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND AIMS Circulating progenitor cells (CPCs) play an important role in vascular repair and can influence cardiovascular (CV) health and longevity. Exercise is known to modulate these cells via mobilization from the bone marrow. The primary aims of this study were to evaluate the association of CPCs with mortality and explore the association between physical activity (PA) and CPCs. METHODS 1751 individuals from the Framingham Offspring cohort (66 ± 9 years [40-92 years], 54% female) were included in the study. CPCs (CD34+, CD34+CD133+, CD34+CD133+KDR+) were measured by flow cytometry. Multivariable Cox regression analyses were performed to investigate relationship of CPCs with future CV event and mortality. Multivariate regression analyses were performed to determine the relationship between self-reported PA and CPC counts. RESULTS Following adjustment for standard risk factors, there was an inverse association between CD34+ CPCs and all-cause mortality (hazard ratio (HR) per unit increase in CD34+, 0.79; 95% CI 0.64-0.98, p = 0.036). CD34+CD133+ CPCs were inversely associated with CV mortality (HR 0.63, 95% CI 0.44-0.91, p = 0.013). Associations of CD34+ and CD34+CD133+ with mortality were strongest in participants with pre-existing CVD. PA was associated with CD34+ CPCs only in CVD participants (PA Index: β = 0.176, p = 0.003; moderate-to-vigorous [MVPA]: β = 0.159, p = 0.007). This relationship was maintained after adjustment for confounding variables. CONCLUSIONS A higher number of CD34+ and CD34+ CD133+ CPCs was inversely associated with all-cause and CV mortality. These associations were strongest in participants with CVD. PA is independently associated with CD34+ CPCs in individuals with CVD only, suggestive of greater benefit for this population group.
Collapse
Affiliation(s)
- David Muggeridge
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, United Kingdom; Institute of Health Research & Innovation, Division of Biomedical Science, University of the Highlands and Islands, Inverness, United Kingdom
| | - Jennifer Dodd
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, United Kingdom
| | - Mark D Ross
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, United Kingdom.
| |
Collapse
|
7
|
Ozcan I, Toya T, Corban MT, Ahmad A, Loeffler D, Morse D, Lerman LO, Kushwaha SS, Lerman A. Circulating Progenitor Cells Are Associated With Plaque Progression And Long-Term Outcomes In Heart Transplant Patients. Cardiovasc Res 2021; 118:1703-1712. [PMID: 34132771 DOI: 10.1093/cvr/cvab203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/10/2021] [Accepted: 06/14/2021] [Indexed: 11/13/2022] Open
Abstract
AIMS Circulating progenitor cells (CPCs) play a role in vascular repair and plaque stability, while osteocalcin (OC) expressing CPCs have been linked to unstable plaque and adverse cardiovascular outcomes. However, their role in cardiac allograft vasculopathy (CAV) has not been elucidated. This cohort study aimed to investigate the contribution of CPCs on CAV progression and cardiovascular events after heart transplantation. METHODS AND RESULTS A total of 80 heart transplant patients (mean age 55 ± 14 years, 72% male) undergoing annual intravascular ultrasound (IVUS) had fresh CPCs marked by CD34, CD133, and OC counted in peripheral blood using flow cytometry, on the same day as baseline IVUS. CAV progression was assessed by IVUS as the change (Δ) in plaque volume divided by segment length (PV/SL), adjusted for the time between IVUS measurements (median 3.0, interquartile range (IQR) [2.8, 3.1] years), and was defined as ΔPV/SL that is above the median ΔPV/SL of study population. Major adverse cardiac events (MACE) was defined as any incident of revascularization, myocardial infarction, heart failure admission, re-transplantation, stroke and death. Patients with higher CD34+CD133+ CPCs had a decreased risk of CAV progression (odds ratio 0.58, 95% confidence interval [CI] [0.37, 0.92], p = 0.01) and MACE (hazard ratio [HR] 0.79, 95% CI [0.66, 0.99], p = 0.05) during a median (IQR) follow up of 8.0 years (7.2, 8.3). Contrarily, higher OC+ cell counts were associated with an increased risk of MACE (HR 1.26, 95% CI [1.03, 1.57], p = 0.02). CONCLUSIONS Lower levels of CD34+CD133+ CPCs are associated with plaque progression and adverse long-term outcomes in patients who underwent allograft heart transplantation. In contrast, higher circulating OC+ levels are associated with adverse long term outcomes. Thus, CPCs might play a role in amelioration of transplant vasculopathy, while OC expression by these cells might play a role in progression. TRANSLATIONAL PERSPECTIVE The results of the current study suggest lower levels of circulating CD34+CD133+ cell levels are associated with cardiac allograft vasculopathy progression and future adverse cardiovascular events, while higher OC+ cell levels are associated with a greater risk of future cardiovascular events. Further studies confirming our findings might elucidate the role of circulating progenitor cells in the pathophysiology of CAV. Moreover, our findings might support the use of circulating progenitors as biomarkers, as well as the notion of cell therapy as potential treatment option for CAV, a disease with severe burden and limited treatment options.
Collapse
Affiliation(s)
- Ilke Ozcan
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Takumi Toya
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Division of Cardiology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Michel T Corban
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ali Ahmad
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Darrell Loeffler
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - David Morse
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Sudhir S Kushwaha
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
8
|
Masson-Meyers DS, Tayebi L. Vascularization strategies in tissue engineering approaches for soft tissue repair. J Tissue Eng Regen Med 2021; 15:747-762. [PMID: 34058083 DOI: 10.1002/term.3225] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/08/2021] [Accepted: 05/17/2021] [Indexed: 12/21/2022]
Abstract
Insufficient vascularization during tissue repair is often associated with poor clinical outcomes. This is a concern especially when patients have critical-sized injuries, where the size of the defect restricts vascularity, or even in small defects that have to be treated under special conditions, such as after radiation therapy (relevant to tumor resection) that hinders vascularity. In fact, poor vascularization is one of the major obstacles for clinical application of tissue engineering methods in soft tissue repair. As a key issue, lack of graft integration, caused by inadequate vascularization after implantation, can lead to graft failure. Moreover, poor vascularization compromises the viability of cells seeded in deep portions of scaffolds/graft materials, due to hypoxia and insufficient nutrient supply. In this article we aim to review vascularization strategies employed in tissue engineering techniques to repair soft tissues. For this purpose, we start by providing a brief overview of the main events during the physiological wound healing process in soft tissues. Then, we discuss how tissue repair can be achieved through tissue engineering, and considerations with regards to the choice of scaffold materials, culture conditions, and vascularization techniques. Next, we highlight the importance of vascularization, along with strategies and methods of prevascularization of soft tissue equivalents, particularly cell-based prevascularization. Lastly, we present a summary of commonly used in vitro methods during the vascularization of tissue-engineered soft tissue constructs.
Collapse
Affiliation(s)
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, USA
| |
Collapse
|
9
|
Khan T, Cabral H. Abnormal Glycosylation of Cancer Stem Cells and Targeting Strategies. Front Oncol 2021; 11:649338. [PMID: 33889547 PMCID: PMC8056457 DOI: 10.3389/fonc.2021.649338] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cell (CSCs) are deemed as one of the main reasons of tumor relapse due to their resistance to standard therapies. Numerous intracellular signaling pathways along with extracellular features are crucial in regulating CSCs properties, such as heterogeneity, plasticity and differentiation. Aberrant glycosylation of these cellular signaling pathways and markers of CSCs have been directly correlated with maintaining survival, self-renewal and extravasation properties. In this review, we highlight the importance of glycosylation in promoting stemness character of CSCs, and present strategies for targeting abnormal glycosylation to eliminate the resistant CSC population.
Collapse
Affiliation(s)
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
10
|
Nandi S, Potunuru UR, Kumari C, Nathan AA, Gopal J, Menon GI, Siddharthan R, Dixit M, Thangaraj PR. Altered kinetics of circulating progenitor cells in cardiopulmonary bypass (CPB) associated vasoplegic patients: A pilot study. PLoS One 2020; 15:e0242375. [PMID: 33211740 PMCID: PMC7676651 DOI: 10.1371/journal.pone.0242375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 10/31/2020] [Indexed: 11/19/2022] Open
Abstract
Vasoplegia observed post cardiopulmonary bypass (CPB) is associated with substantial morbidity, multiple organ failure and mortality. Circulating counts of hematopoietic stem cells (HSCs) and endothelial progenitor cells (EPC) are potential markers of neo-vascularization and vascular repair. However, the significance of changes in the circulating levels of these progenitors in perioperative CPB, and their association with post-CPB vasoplegia, are currently unexplored. We enumerated HSC and EPC counts, via flow cytometry, at different time-points during CPB in 19 individuals who underwent elective cardiac surgery. These 19 individuals were categorized into two groups based on severity of post-operative vasoplegia, a clinically insignificant vasoplegic Group 1 (G1) and a clinically significant vasoplegic Group 2 (G2). Differential changes in progenitor cell counts during different stages of surgery were compared across these two groups. Machine-learning classifiers (logistic regression and gradient boosting) were employed to determine if differential changes in progenitor counts could aid the classification of individuals into these groups. Enumerating progenitor cells revealed an early and significant increase in the circulating counts of CD34+ and CD34+CD133+ hematopoietic stem cells (HSC) in G1 individuals, while these counts were attenuated in G2 individuals. Additionally, EPCs (CD34+VEGFR2+) were lower in G2 individuals compared to G1. Gradient boosting outperformed logistic regression in assessing the vasoplegia grouping based on the fold change in circulating CD 34+ levels. Our findings indicate that a lack of early response of CD34+ cells and CD34+CD133+ HSCs might serve as an early marker for development of clinically significant vasoplegia after CPB.
Collapse
Affiliation(s)
- Sanhita Nandi
- Laboratory of Vascular Biology, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Uma Rani Potunuru
- Apollo Hospitals Educational and Research Foundation, Chennai, India
| | | | - Abel Arul Nathan
- Laboratory of Vascular Biology, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Jayashree Gopal
- Department of Endocrinology and Diabetology, Apollo Hospitals, Chennai, India
- * E-mail: (JG); (MD); (PRT)
| | - Gautam I. Menon
- The Institute of Mathematical Sciences (HBNI), Chennai, India
- Departments of Physics and Biology, Ashoka University, Sonepat, India
| | | | - Madhulika Dixit
- Laboratory of Vascular Biology, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
- * E-mail: (JG); (MD); (PRT)
| | - Paul Ramesh Thangaraj
- Department of Cardiothoracic Surgery, Apollo Hospitals, Chennai, India
- Department of Mechanical Engineering, Indian Institute of Technology Madras, Chennai, India
- * E-mail: (JG); (MD); (PRT)
| |
Collapse
|
11
|
Expression of Pro-Angiogenic Markers Is Enhanced by Blue Light in Human RPE Cells. Antioxidants (Basel) 2020; 9:antiox9111154. [PMID: 33233546 PMCID: PMC7699675 DOI: 10.3390/antiox9111154] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/22/2022] Open
Abstract
Inherited retinal dystrophies are characterized by photoreceptor death. Oxidative stress usually occurs, increasing vision loss, and oxidative damage is often reported in retinitis pigmentosa (RP). More than 300 genes have been reported as RP causing. In contrast, choroidal neovascularization (CNV) only occasionally develops in the late stages of RP. We herein study the regulation of RP causative genes that are likely linked to CNV onset under oxidative conditions. We studied how the endogenous adduct N-retinylidene-N-retinylethanolamine (A2E) affects the expression of angiogenic markers in human retinal pigment epithelium (H-RPE) cells and a possible correlation with RP-causing genes. H-RPE cells were exposed to A2E and blue light for 3 and 6h. By transcriptome analysis, genes differentially expressed between A2E-treated cells and untreated ones were detected. The quantification of differential gene expression was performed by the Limma R package. Enrichment pathway analysis by the FunRich tool and gene prioritization by ToppGene allowed us to identify dysregulated genes involved in angiogenesis and linked to RP development. Two RP causative genes, AHR and ROM1, can be associated with an increased risk of CNV development. Genetic analysis of RP patients affected by CNV will confirm this hypothesis.
Collapse
|
12
|
Kale VP. Transforming growth factor-β boosts the functionality of human bone marrow-derived mesenchymal stromal cells. Cell Biol Int 2020; 44:2293-2306. [PMID: 32749730 DOI: 10.1002/cbin.11437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/17/2020] [Accepted: 08/02/2020] [Indexed: 12/19/2022]
Abstract
Transforming growth factor β1 (TGFβ1) is a negative regulator of hematopoiesis, and yet, it is frequently found at the active sites of hematopoiesis. Here, we show for the first time that bone marrow-derived mononuclear cells (BM MNCs) secrete TGFβ1 in response to erythropoietin (EPO). We further show that human bone marrow-derived mesenchymal stromal cells (BMSCs) briefly exposed to the conditioned medium of EPO-primed MNCs, or purified TGFβ1, gain significantly increased hematopoiesis-supportive ability. Mechanistically, we show that this phenomenon involves TGFβ1-mediated activation of nitric oxide (NO) signalling pathway in the BMSCs. The data suggest that EPO-MNC-TGFβ1 could be one of the regulatory axes operative in the bone marrow microenvironment involved in maintaining the functionality of the resident BMSCs.
Collapse
Affiliation(s)
- Vaijayanti P Kale
- Symbiosis Centre for Stem Cell Research, Symbiosis International University, Pune, India
| |
Collapse
|
13
|
Jasmine S, Thangavelu A, Krishnamoorthy R, Alshatwi AA. Platelet Concentrates as Biomaterials in Tissue Engineering: a Review. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2020. [DOI: 10.1007/s40883-020-00165-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
14
|
Neurovascular Dysfunction in Alzheimer Disease: Assessment of Cerebral Vasoreactivity by Ultrasound Techniques and Evaluation of Circulating Progenitor Cells and Inflammatory Markers. Alzheimer Dis Assoc Disord 2020; 33:212-219. [PMID: 31335454 DOI: 10.1097/wad.0000000000000331] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
AIMS The aims of this study were to assess vascular dysfunction in patients with Alzheimer disease (AD) by investigating cerebral vasomotor reactivity using transcranial Doppler ultrasound (TCD) and to evaluate any correlations between cerebral vasoreactivity and endothelium dysfunction. Moreover, the frequency of circulating progenitor cells (CPCs) and the blood concentration of vascular/inflammatory markers were evaluated. MATERIALS AND METHODS We recruited 35 AD subjects and 17 age-matched, sex-matched, and education-matched healthy control subjects. Cerebral vasomotor reactivity was assessed by means of the TCD-based breath-holding index test (BHI). The level of CPCs was evaluated by means of flow cytometry from venous blood samples, while blood vascular/inflammatory markers were measured by means of enzyme-linked immunosorbent assay. RESULTS Both cerebral assay blood flow velocity in the middle cerebral artery (MCAFV) and BHI values were significantly lower in AD subjects than in healthy controls (P<0.05). A positive trend was found between MCAFV and BHI values and Mini-Mental State Evaluation (MMSE) scores. Moreover, the hematopoietic progenitor cells' count was found to be lower in patients with AD than in controls (P<0.05). Finally, a significantly higher expression of the plasma chemokine CCL-2 was observed in AD patients than in healthy controls. CONCLUSIONS Our results confirm that cerebral hemodynamic deterioration may be a critical marker of cognitive decline. Further studies are needed to investigate the role of circulating CPCs and chemokines as potential contributors to neurovascular dysfunction.
Collapse
|
15
|
Barre A, Naudot M, Colin F, Sevestre H, Collet L, Devauchelle B, Lack S, Marolleau JP, Le Ricousse S. An Alginate-Based Hydrogel with a High Angiogenic Capacity and a High Osteogenic Potential. Biores Open Access 2020; 9:174-182. [PMID: 32642332 PMCID: PMC7337169 DOI: 10.1089/biores.2020.0010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2020] [Indexed: 12/22/2022] Open
Abstract
In bone tissue engineering, autologous cells are combined with osteoconductive scaffolds and implanted into bone defects. The major challenge is the lack of post-implantation vascular growth into biomaterial. The objective of the present study was to develop a new alginate-based hydrogel that enhances the regeneration of bone defects after surgery. The viability of human bone marrow-derived mesenchymal stem cells (BM-MSCs) or human endothelial cells (ECs) cultured alone or together on the hydrogel was analyzed for 24 and 96 h. After seeding, the cells self-assembled and aggregated to form clusters. For functional validation, empty or cellularized hydrogel matrices were implanted ectopically at subcutaneous sites in nude mice. After 2 months, the matrices were explanted. Transplanted human cells were present, and we observed vessels expressing human von Willebrand factor (resulting from the incorporation of transplanted ECs into neovessels and/or the differentiation of BM-MSCs into ECs). The addition of BM-MSCs improved host vascularization and neovessel formation from human cells, relative to ECs alone. Although we did not observe bone formation, the transplanted BM-MSCs were able to differentiate into osteoblasts. This new biomaterial provided an appropriate three-dimensional environment for transplanted cells and has a high angiogenic capacity and an osteogenic potential.
Collapse
Affiliation(s)
- Anaïs Barre
- EA7516, CHIMERE, Jules Verne University of Picardie, Amiens, France
| | - Marie Naudot
- EA7516, CHIMERE, Jules Verne University of Picardie, Amiens, France
| | | | - Henri Sevestre
- Department of Pathology and Anatomy, Amiens University Medical Center, Amiens, France
| | - Louison Collet
- EA4666 HEMATIM, Jules Verne University of Picardie, Amiens, France
| | - Bernard Devauchelle
- EA7516, CHIMERE, Jules Verne University of Picardie, Amiens, France.,Department of Maxillofacial Surgery, Amiens University Medical Center, Amiens, France.,Facing Faces Institute, Amiens, France
| | | | - Jean-Pierre Marolleau
- EA4666 HEMATIM, Jules Verne University of Picardie, Amiens, France.,Facing Faces Institute, Amiens, France.,Department of Hematology, Amiens University Medical Center, Amiens, France
| | - Sophie Le Ricousse
- EA7516, CHIMERE, Jules Verne University of Picardie, Amiens, France.,Facing Faces Institute, Amiens, France
| |
Collapse
|
16
|
Xing Z, Zhao C, Liu H, Fan Y. Endothelial Progenitor Cell-Derived Extracellular Vesicles: A Novel Candidate for Regenerative Medicine and Disease Treatment. Adv Healthc Mater 2020; 9:e2000255. [PMID: 32378361 DOI: 10.1002/adhm.202000255] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 04/12/2020] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of membranous structures, which can be secreted by most cell types. As a product of paracrine secretion, EVs are considered to be a regulatory mediator for intercellular communication. There are many bioactive cargos in EVs, such as proteins, lipids, and nucleic acids. As the precursor cell of vascular endothelial cells (ECs), endothelial progenitor cells (EPCs) are first discovered in peripheral blood. With the development of studies about the functions of EPCs, an increasing number of researchers focus on EPC-derived EVs (EPC-EVs). EPC-EVs exert key functions for promoting angiogenesis in regenerative medicine and show significant therapeutic effects on a variety of diseases such as circulatory diseases, kidney diseases, diabetes, bone diseases, and tissue/organ damages. This article reviews the current knowledge on the role of EPC-EVs in regenerative medicine and disease treatment, discussing the main challenges and future directions in this field.
Collapse
Affiliation(s)
- Zheng Xing
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of EducationSchool of Biological Science and Medical EngineeringBeihang University Beijing 100191 P. R. China
| | - Chen Zhao
- School of Pharmaceutical SciencesTsinghua University Beijing 100084 P. R. China
| | - Haifeng Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of EducationSchool of Biological Science and Medical EngineeringBeihang University Beijing 100191 P. R. China
- Beijing Advanced Innovation Centre for Biomedical EngineeringBeihang University Beijing 100191 P. R. China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of EducationSchool of Biological Science and Medical EngineeringBeihang University Beijing 100191 P. R. China
- Beijing Advanced Innovation Centre for Biomedical EngineeringBeihang University Beijing 100191 P. R. China
- National Research Center for Rehabilitation Technical Aids Beijing 100176 P. R. China
| |
Collapse
|
17
|
Kremer A, Wußmann M, Herrmann M, Raghunath M, Walles H. Ciclopirox olamine promotes the angiogenic response of endothelial cells and mesenchymal stem cells. Clin Hemorheol Microcirc 2020; 73:317-328. [PMID: 31006674 DOI: 10.3233/ch-190559] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Prolyl hydroxylase inhibitors (PHIs) are promising compounds to promote angiogenesis by stabilizing hypoxia-inducible factor-1α (HIF-1α), a master regulator of angiogenesis. Increased HIF-1α presence induces expression of proangiogenic genes such as vascular endothelial growth factor (VEGF). OBJECTIVE We investigated the pharmacological induction of hypoxia via the PHI ciclopirox olamine (CPX) as angiogenesis strategy on human dermal microvascular endothelial cell (hd-mvEC) spheroids directly and indirectly via activating human mesenchymal stem cells (hMSCs). METHODS HMSCs were isolated from bone marrow and hd-mvECs from foreskin biopsies. MSC-conditioned medium after CPX stimulation (MSC-CM CPX) was analyzed by VEGF ELISA and Proteome Profiler™ Human Angiogenesis Array. Direct stimulation with CPX and indirect stimulation via MSC-CM CPX were compared in sprouting assays of hd-mvEC spheroids. RESULTS Direct stimulation with CPX significantly increased sprouting of hd-mvEC spheroids. MSC-CM CPX also induced sprouting from hd-mvEC spheroids, which was mediated by angiogenic VEGF and other proangiogenic factors that had been produced by stimulated hMSCs. CONCLUSIONS The stimulation with CPX increased the proangiogenic response of hd-mvECs and hMSCs. The direct stimulation of hd-mvECs with CPX has the potential to replace external VEGF supplementation. Thus, CPX can induce angiogenesis in ECs even in the absence of auxiliary cells demonstrating a promising proangiogenic approach.
Collapse
Affiliation(s)
- Antje Kremer
- Department Tissue Engineering and Regenerative Medicine (TERM), University Hospital Wuerzburg, Wuerzburg, Germany
| | - Maximiliane Wußmann
- Fraunhofer Translational Center Regenerative Therapies TLC-RT, Fraunhofer Institute for Silicate Research ISC, Wuerzburg, Germany
| | - Marietta Herrmann
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, Wuerzburg, Germany.,Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Michael Raghunath
- Institute of Chemistry and Biotechnology, Zuerich University of Applied Sciences (ZHAW), Waedenswil, Switzerland.,Competence Center Tissue Engineering for Drug Discover (TEDD), ZHAW, Waedenswil, Switzerland
| | - Heike Walles
- Fraunhofer Translational Center Regenerative Therapies TLC-RT, Fraunhofer Institute for Silicate Research ISC, Wuerzburg, Germany
| |
Collapse
|
18
|
Kashiwazaki D, Koh M, Uchino H, Akioka N, Kuwayama N, Noguchi K, Kuroda S. Hypoxia accelerates intraplaque neovascularization derived from endothelial progenitor cells in carotid stenosis. J Neurosurg 2019; 131:884-891. [PMID: 30485214 DOI: 10.3171/2018.4.jns172876] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 04/09/2018] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The relationship between intraplaque hypoxia and intraplaque hemorrhage (IPH) has been reported, but the details remain obscure. In this study, the authors aimed to clarify the relationship among intraplaque hypoxia, endothelial progenitor cells (EPCs), and neovascularization, which causes IPH. The histological findings of specimens obtained from carotid endarterectomy were assessed. METHODS This study included 49 patients who underwent carotid endarterectomy. Magnetic resonance plaque imaging was performed to analyze the components of the carotid plaques, and surgical specimens were subjected to immunohistochemical analysis. The numbers of hypoxia-inducible factor-1 alpha (HIF-1α)-, CD34-, CD133-, and vascular endothelial growth factor receptor-2 (VEGFR-2)-positive cells in the carotid plaques were precisely quantified, as were the number and maximum diameter of CD31-positive microvessels. RESULTS Plaque components were judged as fibrous in 7 samples, lipid-rich in 22, and IPH in 20. The number of CD34-, VEGFR-2-, and CD133-positive cells as an EPC-specific marker was significantly correlated with the number of HIF-1α-positive cells (r = 0.9, r = 0.82, and r = 0.81, respectively). These numbers varied among the 3 plaque components (IPH > lipid-rich > fibrous). The number and maximum luminal diameter of CD31-positive microvessels were also significantly correlated with the number of HIF-1α-positive cells (r = 0.85 and r = 0.89, respectively) and varied among the 3 plaque components (IPH > lipid-rich > fibrous). CONCLUSIONS The present findings suggest that intraplaque hypoxia may accelerate abnormal microvessel formation derived from EPCs, which in turn promotes IPH. The results also suggest that microvessel enlargement is a pivotal characteristic of IPH and these enlarged microvessels are immature endothelial tubes with disorganized branching and are fragile and prone to rupture.
Collapse
Affiliation(s)
| | | | | | | | | | - Kyo Noguchi
- 2Radiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Japan
| | | |
Collapse
|
19
|
Phenotypic Characterization of Bone Marrow Mononuclear Cells and Derived Stromal Cell Populations from Human Iliac Crest, Vertebral Body and Femoral Head. Int J Mol Sci 2019; 20:ijms20143454. [PMID: 31337109 PMCID: PMC6678175 DOI: 10.3390/ijms20143454] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 12/28/2022] Open
Abstract
(1) In vitro, bone marrow-derived stromal cells (BMSCs) demonstrate inter-donor phenotypic variability, which presents challenges for the development of regenerative therapies. Here, we investigated whether the frequency of putative BMSC sub-populations within the freshly isolated mononuclear cell fraction of bone marrow is phenotypically predictive for the in vitro derived stromal cell culture. (2) Vertebral body, iliac crest, and femoral head bone marrow were acquired from 33 patients (10 female and 23 male, age range 14–91). BMSC sub-populations were identified within freshly isolated mononuclear cell fractions based on cell-surface marker profiles. Stromal cells were expanded in monolayer on tissue culture plastic. Phenotypic assessment of in vitro derived cell cultures was performed by examining growth kinetics, chondrogenic, osteogenic, and adipogenic differentiation. (3) Gender, donor age, and anatomical site were neither predictive for the total yield nor the population doubling time of in vitro derived BMSC cultures. The abundance of freshly isolated progenitor sub-populations (CD45−CD34−CD73+, CD45−CD34−CD146+, NG2+CD146+) was not phenotypically predictive of derived stromal cell cultures in terms of growth kinetics nor plasticity. BMSCs derived from iliac crest and vertebral body bone marrow were more responsive to chondrogenic induction, forming superior cartilaginous tissue in vitro, compared to those isolated from femoral head. (4) The identification of discrete progenitor populations in bone marrow by current cell-surface marker profiling is not predictive for subsequently derived in vitro BMSC cultures. Overall, the iliac crest and the vertebral body offer a more reliable tissue source of stromal progenitor cells for cartilage repair strategies compared to femoral head.
Collapse
|
20
|
Herrmann M, Stanić B, Hildebrand M, Alini M, Verrier S. In vitro simulation of the early proinflammatory phase in fracture healing reveals strong immunomodulatory effects of CD146-positive mesenchymal stromal cells. J Tissue Eng Regen Med 2019; 13:1466-1481. [PMID: 31132812 DOI: 10.1002/term.2902] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 03/27/2019] [Accepted: 04/29/2019] [Indexed: 01/06/2023]
Abstract
The impact of microenvironmental cues and changes due to injury on the phenotype and fate of mesenchymal stromal cells (MSCs) is poorly understood. Here, we aimed to simulate the microenvironment associated with the early stage of bone healing in vitro and to study the regenerative response of MSCs. We enriched CD146+ MSCs from the human bone marrow. Different physiological and pathological microenvironments were simulated by using conditioned medium (CM) from human endothelial cells and osteoblasts (healthy bone), femoral head-derived bone fragments (injured bone), and activated platelets (platelet-rich plasma [PRP], injury). Cells were incubated in CM and analyzed with respect to proliferation, gene expression, migration, osteogenic differentiation, and their effect on polyclonally induced proliferation of peripheral blood mononuclear cells. CD146+ MSCs showed a specific response to different microenvironments. Cell proliferation was observed in all media with the highest values in PRP-CM and injured bone-CM. Gene expression analysis revealed the upregulation of chemokines, proinflammatory, proangiogenic, and genes involved in immunomodulation in cells stimulated with PRP- and injured bone-CM, suggesting strong paracrine activity. PRP-CM led to pronounced inhibition of lymphocyte proliferation by CD146+MSCs. Our results indicate that a microenvironment simulating bone injury elicits strong immunomodulatory and proangiogenic activity of CD146+ MSCs. This suggests that in the early stage of bone healing, the prime function of MSCs and their CD146+ subpopulation is in regulating the immune response and inducing neovascularization. Future studies will investigate the key components in CM driving this function, which might be potential targets to therapeutically stimulate the regenerative potential of MSCs.
Collapse
Affiliation(s)
- Marietta Herrmann
- AO Research Institute Davos, Davos Platz, Switzerland.,IZKF Research Group Tissue Regeneration in Musculoskeletal Diseases, University Clinics Würzburg and Orthopedic Center for Musculoskeletal Research, University of Würzburg, Germany
| | | | | | - Mauro Alini
- AO Research Institute Davos, Davos Platz, Switzerland
| | | |
Collapse
|
21
|
Pereira RC, Benelli R, Canciani B, Scaranari M, Daculsi G, Cancedda R, Gentili C. Beta-tricalcium phosphate ceramic triggers fast and robust bone formation by human mesenchymal stem cells. J Tissue Eng Regen Med 2019; 13:1007-1018. [PMID: 30811859 DOI: 10.1002/term.2848] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 02/14/2019] [Accepted: 02/21/2019] [Indexed: 12/13/2022]
Abstract
Due to their osteoconductive and inductive properties, a variety of calcium phosphate (CaP) scaffolds are commonly used in orthopaedics as graft material to heal bone defects. In this study, we have used two CaP scaffolds with different hydroxyapatite (HA) and β-tricalcium phosphate (β-TCP) ratios (MBCP®; 60/40 and MBCP+ ®; 20/80) to investigate their intrinsic capacity to favour human bone marrow stem cells (hBMSCs) osteogenic differentiation capacity. We report that MBCP+ ® showed in in vitro culture model a higher rate of calcium ion release in comparison with MBCP®. In two defined coculture systems, the hBMSC seeded onto MBCP+ ® presented an increased amount of VEGF secretion, resulting in an enhanced endothelial cell proliferation and capillary formation compared with hBMSC seeded onto MBCP®. When both ceramics combined with hBMSC were implanted in a nude mouse model, we observed a faster osteogenic differentiation and enhancement mature bone deposition sustained by the presence of a vast host vasculature within the MBCP+ ® ceramics. Bone formation was observed in samples highly positive to the activation of calcium sensing receptor protein (CaSr) on the surface of seeded hBMSC that also shown higher BMP-2 protein expression. With these data we provide valuable insights in the possible mechanisms of ossification and angiogenesis by hBMSC that we believe to be primed by calcium ions released from CaP scaffolds. Evidences could lead to an optimization of ceramic scaffolds to prime bone repair.
Collapse
Affiliation(s)
- Rui C Pereira
- Laboratory of Regenerative Medicine, Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Roberto Benelli
- Laboratory of Immunology, IRCCS AOU San Martino, Genoa, Italy
| | - Barbara Canciani
- Laboratory of Regenerative Medicine, Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Monica Scaranari
- Laboratory of Regenerative Medicine, Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Guy Daculsi
- INSERM LIOAD U791, Dental Faculty, Nantes University, Nantes, France
| | - Ranieri Cancedda
- Laboratory of Regenerative Medicine, Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Chiara Gentili
- Laboratory of Regenerative Medicine, Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy.,Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| |
Collapse
|
22
|
Isolation and characterization of endothelial colony-forming cells from mononuclear cells of rat bone marrow. Exp Cell Res 2018; 370:116-126. [PMID: 29908162 DOI: 10.1016/j.yexcr.2018.06.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 06/12/2018] [Accepted: 06/13/2018] [Indexed: 12/25/2022]
Abstract
Transplantation of bone marrow-derived endothelial progenitor cells (BM-EPCs) has been used as a therapeutic strategy for vascular repair. However, it remains controversial whether BM-EPCs exhibit clonal endothelial colony-forming cell (ECFC) capacity, a characteristic of true EPCs. The aim of this study was to isolate and explore the cellular properties of BM-ECFCs. We isolated BM-ECFCs from rat bone marrow with high purity via an optimized method. This approach involved the removal of selective colonies based on the conventional differential adhesive culture method used to isolate ECFCs from peripheral and umbilical cord blood. Our results indicate that primary colony BM-ECFCs display a panel of surface antigen markers consistent with endothelial cells. These BM-ECFCs coexpress CD34, CD133, and VEGFR2 at high levels, and these levels decrease with passaging. These cells have high potential for proliferation, migration, and formation of capillary-like structures on Matrigel, and these abilities are retained during ex vivo expansion. Furthermore, BM-ECFCs cultured with 10% or 20% fetal bovine serum demonstrated two different patterns of spontaneous capillary-like structure formation. These results provide a foundation for isolation of ECFCs from human bone marrow for autologous cell transplantation and tissue engineering applications in the future.
Collapse
|
23
|
VCAM-1 expression is upregulated by CD34+/CD133+-stem cells derived from septic patients. PLoS One 2018; 13:e0195064. [PMID: 29601599 PMCID: PMC5877884 DOI: 10.1371/journal.pone.0195064] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 03/15/2018] [Indexed: 12/29/2022] Open
Abstract
CD34+/CD133+- cells are a bone marrow derived stem cell population, which presumably contain vascular progenitor cells and are associated with improved vascular repair. In this study, we investigated whether the adhesion molecules ICAM-1 (intercellular adhesion molecule-1), VCAM-1 (vascular adhesion molecule-1), E-selectin und L-selectin, which are involved in homing of vascular stem cells, are upregulated by CD34+/CD133+-stem cells from septic patients and would be associated with improved clinical outcome. Peripheral blood mononuclear cells from intensive care unit (ICU) patients with (n = 30) and without sepsis (n = 10), and healthy volunteers (n = 15) were isolated using Ficoll density gradient centrifugation. The expression of VCAM-1, ICAM-1, E-selectin and L-selectin was detected on CD34+/CD133+-stem cells by flow cytometry. The severity of disease was assessed by the Simplified Acute Physiology Score (SAPS) II. Serum concentrations of vascular endothelial growth factor (VEGF) and angiopoietin (Ang)-2 were determined by Enzyme-linked immunosorbent assay. The expression of VCAM-1, ICAM-1, E-selectin and L-selectin by CD34+/CD133+-stem cells was significantly upregulated in septic patients, and correlated with sepsis severity. Furthermore, high expression of VCAM-1 by CD34+/CD133+-stem cells revealed a positive association with mortalitiy (p<0.05). Furthermore, significantly higher serum concentrations of VEGF and Ang-2 were found in septic patients, however none showed a strong association with survival. Our data suggest, that VCAM-1 upregulation on CD34+/CD133+-stem cells could play a crucial role in their homing in the course of sepsis. An increase in sepsis severity resulted in both and increase in CD34+/CD133+-stem cells and VCAM-1-expression by those cells, which might reflect an increase in need for vascular repair.
Collapse
|
24
|
Naskou MC, Sumner SM, Chocallo A, Kemelmakher H, Thoresen M, Copland I, Galipeau J, Peroni JF. Platelet lysate as a novel serum-free media supplement for the culture of equine bone marrow-derived mesenchymal stem cells. Stem Cell Res Ther 2018; 9:75. [PMID: 29566772 PMCID: PMC5863827 DOI: 10.1186/s13287-018-0823-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 03/01/2018] [Indexed: 12/19/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) produced for clinical purposes rely on culture media containing fetal bovine serum (FBS) which is xenogeneic and has the potential to significantly alter the MSC phenotype, rendering these cells immunogenic. As a result of bovine-derived exogenous proteins expressed on the cell surface, MSCs may be recognized by the host immune system as non-self and be rejected. Platelet lysate (PL) may obviate some of these concerns and shows promising results in human medicine as a possible alternative to FBS. Our goal was to evaluate the use of equine platelet lysate (ePL) pooled from donor horses in place of FBS to culture equine MSCs. We hypothesized that ePL, produced following apheresis, will function as the sole media supplement to accelerate the expansion of equine bone marrow-derived MSCs without altering their phenotype and their immunomodulatory capacity. Methods Platelet concentrate was obtained via plateletpheresis and ePL were produced via freeze-thaw and centrifugation cycles. Population doublings (PD) and doubling time (DT) of bone marrow-derived MSCs (n = 3) cultured with FBS or ePL media were calculated. Cell viability, immunophenotypic analysis, and trilineage differentiation capacity of MSCs were assessed accordingly. To assess the ability of MSCs to modulate inflammatory responses, E. coli lipopolysaccharide (LPS)-stimulated monocytes were cocultured with MSCs cultured in the two different media formulations, and cell culture supernatants were assayed for the production of tumor necrosis factor (TNF)-α. Results Our results showed that MSCs cultured in ePL media exhibited similar proliferation rates (PD and DT) compared with those cultured in FBS at individual time points. MSCs cultured in ePL showed a statistically significant increased viability following a single washing step, expressed similar levels of MSC markers compared to FBS, and were able to differentiate towards the three lineages. Finally, MSCs cultured in ePL efficiently suppressed the release of TNF-α when exposed to LPS-stimulated monocytes similar to those cultured in FBS. Conclusion ePL has the potential to be used for the expansion of MSCs before clinical application, avoiding the concerns associated with the use of FBS.
Collapse
Affiliation(s)
- Maria C Naskou
- Department of Large Animal Medicine, Veterinary Medical Center, College of Veterinary Medicine, University of Georgia, 2200 College Station Road, Athens, GA, 30602, USA
| | - Scarlett M Sumner
- Department of Large Animal Medicine, Veterinary Medical Center, College of Veterinary Medicine, University of Georgia, 2200 College Station Road, Athens, GA, 30602, USA
| | - Anna Chocallo
- Department of Large Animal Medicine, Veterinary Medical Center, College of Veterinary Medicine, University of Georgia, 2200 College Station Road, Athens, GA, 30602, USA
| | - Hannah Kemelmakher
- Department of Large Animal Medicine, Veterinary Medical Center, College of Veterinary Medicine, University of Georgia, 2200 College Station Road, Athens, GA, 30602, USA
| | - Merrilee Thoresen
- Department of Large Animal Medicine, Veterinary Medical Center, College of Veterinary Medicine, University of Georgia, 2200 College Station Road, Athens, GA, 30602, USA
| | - Ian Copland
- Emory Personalized Immunotherapy Center [EPIC], Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA, 30322, USA
| | - Jacques Galipeau
- Department of Medicine and Carbone Comprehensive Cancer Center, University of Wisconsin, 600 Highland Ave., Madison, WI, 53792, USA
| | - John F Peroni
- Department of Large Animal Medicine, Veterinary Medical Center, College of Veterinary Medicine, University of Georgia, 2200 College Station Road, Athens, GA, 30602, USA.
| |
Collapse
|
25
|
Herrmann M, Zeiter S, Eberli U, Hildebrand M, Camenisch K, Menzel U, Alini M, Verrier S, Stadelmann VA. Five Days Granulocyte Colony-Stimulating Factor Treatment Increases Bone Formation and Reduces Gap Size of a Rat Segmental Bone Defect: A Pilot Study. Front Bioeng Biotechnol 2018; 6:5. [PMID: 29484293 PMCID: PMC5816045 DOI: 10.3389/fbioe.2018.00005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/15/2018] [Indexed: 12/25/2022] Open
Abstract
Bone is an organ with high natural regenerative capacity and most fractures heal spontaneously when appropriate fracture fixation is provided. However, additional treatment is required for patients with large segmental defects exceeding the endogenous healing potential and for patients suffering from fracture non-unions. These cases are often associated with insufficient vascularization. Transplantation of CD34+ endothelial progenitor cells (EPCs) has been successfully applied to promote neovascularization of bone defects, however including extensive ex vivo manipulation of cells. Here, we hypothesized, that treatment with granulocyte colony-stimulating factor (G-CSF) may improve bone healing by mobilization of CD34+ progenitor cells into the circulation, which in turn may facilitate vascularization at the defect site. In this pilot study, we aimed to characterize the different cell populations mobilized by G-CSF and investigate the influence of cell mobilization on the healing of a critical size femoral defect in rats. Cell mobilization was investigated by flow cytometry at different time points after five consecutive daily G-CSF injections. In a pilot study, bone healing of a 4.5-mm critical femoral defect in F344 rats was compared between a saline-treated control group and a G-CSF treatment group. In vivo microcomputed tomography and histology were applied to compare bone formation in both treatment groups. Our data revealed that leukocyte counts show a peak increase at the first day after the last G-CSF injection. In addition, we found that CD34+ progenitor cells, including EPCs, were significantly enriched at day 1, and further increased at day 5 and day 11. Upregulation of monocytes, granulocytes and macrophages peaked at day 1. G-CSF treatment significantly increased bone volume and bone density in the defect, which was confirmed by histology. Our data show that different cell populations are mobilized by G-CSF treatment in cell specific patterns. Although in this pilot study no bridging of the critical defect was observed, significantly improved bone formation by G-CSF treatment was clearly shown.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland
| | | | | |
Collapse
|
26
|
Nation DA, Tan A, Dutt S, McIntosh EC, Yew B, Ho JK, Blanken AE, Jang JY, Rodgers KE, Gaubert A. Circulating Progenitor Cells Correlate with Memory, Posterior Cortical Thickness, and Hippocampal Perfusion. J Alzheimers Dis 2018; 61:91-101. [PMID: 29103037 PMCID: PMC5924766 DOI: 10.3233/jad-170587] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Bone marrow-derived progenitor cells survey the vasculature and home to sites of tissue injury where they can promote repair and regeneration. It has been hypothesized that these cells may play a protective role neurodegenerative and vascular cognitive impairment. OBJECTIVE To evaluate progenitor cell levels in older adults with and without mild cognitive impairment (MCI), and to relate circulating levels to memory, brain volume, white matter lesion volume, and cerebral perfusion. METHOD Thirty-two older adults, free of stroke and cardiovascular disease, were recruited from the community and evaluated for diagnosis of MCI versus cognitively normal (CN). Participants underwent brain MRI and blood samples were taken to quantify progenitor reserve using flow cytometry (CD34+, CD34+CD133+, and CD34+CD133+CD309+ cells). RESULTS Participants with MCI (n = 10) exhibited depletion of all CPC markers relative to those who were CN (n = 22), after controlling for age, sex, and education. Post-hoc age, sex, and education matched comparisons (n = 10 MCI, n = 10 CN) also revealed the same pattern of results. Depletion of CD34+ cells correlated with memory performance, left posterior cortical thickness, and bilateral hippocampal perfusion. Participants exhibited low levels of vascular risk and white matter lesion burden that did not correlate with progenitor levels. CONCLUSIONS Circulating progenitor cells are associated with cognitive impairment, memory, cortical atrophy, and hippocampal perfusion. We hypothesize that progenitor depletion contributes to, or is triggered by, cognitive decline and cortical atrophy. Further study of progenitor cell depletion in older adults may benefit efforts to prevent or delay dementia.
Collapse
Affiliation(s)
- Daniel A. Nation
- Department of Psychology, University of Southern California, Los Angeles, CA
| | - Alick Tan
- Department of Clinical Pharmacy, University of Southern California, Los Angeles CA
| | - Shubir Dutt
- Department of Psychology, University of Southern California, Los Angeles, CA
| | - Elissa C. McIntosh
- Department of Psychology, University of Southern California, Los Angeles, CA
| | - Belinda Yew
- Department of Psychology, University of Southern California, Los Angeles, CA
| | - Jean K. Ho
- Department of Psychology, University of Southern California, Los Angeles, CA
| | - Anna E. Blanken
- Department of Psychology, University of Southern California, Los Angeles, CA
| | - Jung Yun Jang
- Department of Psychology, University of Southern California, Los Angeles, CA
| | - Kathleen E. Rodgers
- Department of Clinical Pharmacy, University of Southern California, Los Angeles CA
| | - Aimée Gaubert
- Department of Psychology, University of Southern California, Los Angeles, CA
| |
Collapse
|
27
|
Sradnick J, Tselmin S, Wagner A, Julius U, Todorov V, Hugo C, Hohenstein B. H.E.L.P apheresis exerts long term effects on the capacity of circulating proangiogenic cells. ATHEROSCLEROSIS SUPP 2017; 30:232-237. [PMID: 29096843 DOI: 10.1016/j.atherosclerosissup.2017.05.045] [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: 11/29/2022]
Abstract
BACKGROUND Severe forms of mono- and polygenetic hypercholesterolemia as well as elevated Lipoprotein (a) (LP(a)) with progressing cardiovascular (CV) disease are indication for lipoprotein apheresis (LA) in Germany. Many studies investigated pleiotropic effects of LA that might contribute to beneficial effects in advanced atherosclerosis. The present study aimed at investigating the potential role of Proangiogenic Cells (PAC) in patients with new onset or chronic LA using the heparin induced extracorporeal LDL-precipitation (H.E.L.P.) apheresis system. METHODS Patients (n = 10) new to LA and HELP treatment were investigated immediately before, shortly after, 24 h later and 4 weeks following LA. Peripheral blood was used to count PAC in circulation via flow cytometry. In a second step, blood cells from patients were cultured in endothelial selective medium and further evaluated for adhesion in fibronectin coated chamber slides and migratory capacity (stromal cell-derived factor-1 (SDF-1) induced migration). RESULTS Cells expressing typical EPC markers were rarely detected in blood samples. No differences occurred over time in CD34+; CD34+ CD133+ CD45-; CD34+/KDR+ and CXCR4+/CD14+ positive PAC. We found no differences in cell adhesion at the different time points, while significantly more cells migrated into the SDF-1 medium following four weeks of continuing apheresis therapy. CONCLUSION Using well established systems, this study was not able to demonstrate relevant acute effects of LA on PAC in patients new to LA. The increased migratory capacity of PAC might be an indicator of chronic beneficial pleiotropic effects in patients undergoing H.E.L.P. apheresis.
Collapse
Affiliation(s)
- Jan Sradnick
- Division of Nephrology, Department of Internal Medicine III, University Hospital and Faculty of Medicine Carl Gustav Carus at the Technische Universität, Dresden, Germany
| | - Sergey Tselmin
- Extracorporeal Treatment and Apheresis Center, Department of Internal Medicine III, University Hospital and Faculty of Medicine Carl Gustav Carus at the Technische Universität, Dresden, Germany
| | - Andrea Wagner
- Division of Nephrology, Department of Internal Medicine III, University Hospital and Faculty of Medicine Carl Gustav Carus at the Technische Universität, Dresden, Germany
| | - Ulrich Julius
- Extracorporeal Treatment and Apheresis Center, Department of Internal Medicine III, University Hospital and Faculty of Medicine Carl Gustav Carus at the Technische Universität, Dresden, Germany
| | - Vladimir Todorov
- Division of Nephrology, Department of Internal Medicine III, University Hospital and Faculty of Medicine Carl Gustav Carus at the Technische Universität, Dresden, Germany
| | - Christian Hugo
- Division of Nephrology, Department of Internal Medicine III, University Hospital and Faculty of Medicine Carl Gustav Carus at the Technische Universität, Dresden, Germany
| | - Bernd Hohenstein
- Division of Nephrology, Department of Internal Medicine III, University Hospital and Faculty of Medicine Carl Gustav Carus at the Technische Universität, Dresden, Germany; Extracorporeal Treatment and Apheresis Center, Department of Internal Medicine III, University Hospital and Faculty of Medicine Carl Gustav Carus at the Technische Universität, Dresden, Germany.
| |
Collapse
|
28
|
Zhang X, Li J, Ye P, Gao G, Hubbell K, Cui X. Coculture of mesenchymal stem cells and endothelial cells enhances host tissue integration and epidermis maturation through AKT activation in gelatin methacryloyl hydrogel-based skin model. Acta Biomater 2017; 59:317-326. [PMID: 28684336 DOI: 10.1016/j.actbio.2017.07.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/26/2017] [Accepted: 07/01/2017] [Indexed: 12/25/2022]
Abstract
A major challenge for clinical use of skin substitutes is insufficient host tissue integration leading to loosening and partial necrosis of the implant. In this present study, a three-dimensional (3D) coculture system constructed using human umbilical cord mesenchymal stem cells (uc-MSCs) and umbilical vein endothelial cells (HUVECs) encapsulated in gelatin methacryloyl (GelMA) hydrogels was evaluated to determine the outcomes of cell-cell interactions in vitro and in vivo. The results revealed that GelMA hydrogels displayed minor cytotoxicity on both cell types. An uc-MSC:HUVEC ratio of 50:50 demonstrated the highest cell proliferation and expression of angiogenic markers. The supplement of basic fibroblast growth factors (bFGF) in coculture system further induced cell proliferation and gene expression in vitro. In vivo transplantation of this cocultured constructs efficiently enhanced the implant and host tissue integration. Additionally, the proliferation of keratinocytes was well maintained on GelMA hydrogels and the gene expression related to cell proliferation and differentiation was significantly increased in coculture system comparing to monoculture. Mechanistically, AKT signaling pathways were activated in cocultures. Our findings suggest that coculturing MSC and EC in GelMA hydrogels could be a promising approach to substantially improve the integration of exogenous skin substitutes and host tissues. STATEMENT OF SIGNIFICANCE In this study, the co-culture of uc-MSCs and HUVECs in photocrosslinkable GelMA hydrogels significantly enhanced host tissue integration. Cell proliferation, ECM deposition and angiogenic genes expression were all substantially improved in vitro and the excellent host tissue integration into the implanted tissue was observed in vivo. When served as a dermal layer, the scaffold with co-cultured cells enhanced the proliferation and differentiation of keratinocytes. AKT signaling was proved to be involved in the regulation of cell survival and fate determination. This work demonstrated the importance of 3D cell co-culture to facilitate host tissue integration that can be a promising approach for long-term survival of skin substitutes.
Collapse
Affiliation(s)
- Xiaofei Zhang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Jun Li
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Pengxiang Ye
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Guifang Gao
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China; Stemorgan Incorporated, Allen, TX, USA.
| | | | - Xiaofeng Cui
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China; Stemorgan Incorporated, Allen, TX, USA.
| |
Collapse
|
29
|
Platelet-Rich Plasma as an Autologous and Proangiogenic Cell Delivery System. Mediators Inflamm 2017; 2017:1075975. [PMID: 28845088 PMCID: PMC5563430 DOI: 10.1155/2017/1075975] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 06/20/2017] [Indexed: 12/18/2022] Open
Abstract
Angiogenesis is a key factor in early stages of wound healing and is crucial for the repair of vascularized tissues such as the bone. However, supporting timely revascularization of the defect site still presents a clinical challenge. Tissue engineering approaches delivering endothelial cells or prevascularized constructs may overcome this problem. In the current study, we investigated platelet-rich plasma (PRP) gels as autologous, injectable cell delivery systems for prevascularized constructs. PRP was produced from human thrombocyte concentrates. GFP-expressing human umbilical vein endothelial cells (HUVECs) and human bone marrow-derived mesenchymal stem cells (MSCs) were encapsulated in PRP gels in different proportions. The formation of cellular networks was assessed over 14 days by time-lapse microscopy, gene expression analysis, and immunohistology. PRP gels presented a favorable environment for the formation of a three-dimensional (3D) cellular network. The formation of these networks was apparent as early as 3 days after seeding. Networks increased in complexity and branching over time but were only stable in HUVEC-MSC cocultures. The high cell viability together with the 3D capillary-like networks observed at early time points suggests that PRP can be used as an autologous and proangiogenic cell delivery system for the repair of vascularized tissues such as the bone.
Collapse
|
30
|
Marzec M, Kucińska-Lipka J, Kalaszczyńska I, Janik H. Development of polyurethanes for bone repair. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 80:736-747. [PMID: 28866223 DOI: 10.1016/j.msec.2017.07.047] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 01/23/2017] [Accepted: 07/29/2017] [Indexed: 12/12/2022]
Abstract
The purpose of this paper is to review recent developments on polyurethanes aimed at the design, synthesis, modifications, and biological properties in the field of bone tissue engineering. Different polyurethane systems are presented and discussed in terms of biodegradation, biocompatibility and bioactivity. A comprehensive discussion is provided of the influence of hard to soft segments ratio, catalysts, stiffness and hydrophilicity of polyurethanes. Interaction with various cells, behavior in vivo and current strategies in enhancing bioactivity of polyurethanes are described. The discussion on the incorporation of biomolecules and growth factors, surface modifications, and obtaining polyurethane-ceramics composites strategies is held. The main emphasis is placed on the progress of polyurethane applications in bone regeneration, including bone void fillers, shape memory scaffolds, and drug carrier.
Collapse
Affiliation(s)
- M Marzec
- Department of Polymer Technology, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
| | - J Kucińska-Lipka
- Department of Polymer Technology, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland.
| | - I Kalaszczyńska
- Department of Histology and Embryology, Center for Biostructure Research, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; Centre for Preclinical Research and Technology, Banacha 1b, 02-097 Warsaw, Poland
| | - H Janik
- Department of Polymer Technology, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
| |
Collapse
|
31
|
Souidi N, Stolk M, Rudeck J, Strunk D, Schallmoser K, Volk HD, Seifert M. Stromal Cells Act as Guardians for Endothelial Progenitors by Reducing Their Immunogenicity After Co-Transplantation. Stem Cells 2017; 35:1233-1245. [PMID: 28100035 DOI: 10.1002/stem.2573] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 12/07/2016] [Accepted: 12/18/2016] [Indexed: 01/15/2023]
Abstract
Regeneration of injured tissues requires effective therapeutic strategies supporting vasculogenesis. The lack of instantly available autologous cell sources and immunogenicity of allogeneic endothelial (progenitor) cells limits clinical progress. Based on the immunosuppressive potency of mesenchymal stem/progenitor cells (MSCs), we investigated whether crosstalk between endothelial colony-forming progenitor cells (ECFCs) and MSCs during vasculogenesis could lower allogeneic T cell responses against ECFCs allowing long-term engraftment in vivo. Immunodeficient mice received subcutaneous grafts containing human ECFCs alone, or pairs of human ECFCs/MSCs from the same umbilical cord (UC) to study vasculogenesis in the presence of human leukocyte antigen (HLA)-mismatched human peripheral blood mononuclear cells (PBMCs). In vitro, cell surface marker changes due to interferon gamma (IFNγ) stimulation during ECFC/MSC coculture were determined and further effects on allostimulated T cell proliferation and cytotoxic lysis were measured. IFNγ-induced HLA-DR expression on ECFCs and MSCs, but both cell types had significantly less HLA-DR in cocultures. ECFC-induced T cell proliferation was abolished after MSC coculture as a result of HLA-DR downregulation and indolamin-2,3-dioxygenase activation. Additionally, allospecific CD8+ T cell-mediated lysis of ECFCs was reduced in cocultures. ECFC/MSC coapplication in immunodeficient mice not only promoted the generation of improved blood vessel architecture after 6 weeks, but also reduced intragraft immune cell infiltration and endothelial HLA-DR expression following PBMC reconstitution. Crosstalk between UC-derived ECFCs and MSCs after combined transplantation can lower the risk of ECFC rejection, thus enabling their coapplication for therapeutic vasculogenesis. Stem Cells 2017;35:1233-1245.
Collapse
Affiliation(s)
- Naima Souidi
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Meaghan Stolk
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Juliane Rudeck
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Dirk Strunk
- Institute of Experimental and Clinical Cell Therapy, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord & Tissue Regeneration Center, Paracelsus Medical University, Salzburg, Austria
| | - Katharina Schallmoser
- Spinal Cord & Tissue Regeneration Center, Paracelsus Medical University, Salzburg, Austria.,Department for Blood Group Serology and Transfusion Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Hans-Dieter Volk
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Martina Seifert
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| |
Collapse
|
32
|
Preparation, physicochemical properties and biocompatibility of PBLG/PLGA/bioglass composite scaffolds. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 71:118-124. [DOI: 10.1016/j.msec.2016.09.085] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/31/2016] [Accepted: 09/29/2016] [Indexed: 12/13/2022]
|
33
|
Måløy M, Måløy F, Jakobsen P, Olav Brandsdal B. Dynamic self-organisation of haematopoiesis and (a)symmetric cell division. J Theor Biol 2017; 414:147-164. [DOI: 10.1016/j.jtbi.2016.11.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 11/06/2016] [Accepted: 11/30/2016] [Indexed: 12/24/2022]
|
34
|
Comparison of Endothelial Differentiation Capacities of Human and Rat Adipose-Derived Stem Cells. Plast Reconstr Surg 2016; 138:1231-1241. [DOI: 10.1097/prs.0000000000002791] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
35
|
Astori G, Amati E, Bambi F, Bernardi M, Chieregato K, Schäfer R, Sella S, Rodeghiero F. Platelet lysate as a substitute for animal serum for the ex-vivo expansion of mesenchymal stem/stromal cells: present and future. Stem Cell Res Ther 2016; 7:93. [PMID: 27411942 PMCID: PMC4944312 DOI: 10.1186/s13287-016-0352-x] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The use of fetal bovine serum (FBS) as a cell culture supplement is discouraged by regulatory authorities to limit the risk of zoonoses and xenogeneic immune reactions in the transplanted host. Additionally, FBS production came under scrutiny due to animal welfare concerns. Platelet derivatives have been proposed as FBS substitutes for the ex-vivo expansion of mesenchymal stem/stromal cells (MSCs) since platelet-derived growth factors can promote MSC ex-vivo expansion. Platelet-derived growth factors are present in platelet lysate (PL) obtained after repeated freezing–thawing cycles of the platelet-rich plasma or by applying physiological stimuli such as thrombin or CaCl2. PL-expanded MSCs have been used already in the clinic, taking advantage of their faster proliferation compared with FBS-expanded preparations. Should PL be applied to other biopharmaceutical products, its demand is likely to increase dramatically. The use of fresh platelet units for the production of PL raises concerns due to limited availability of platelet donors. Expired units might represent an alternative, but further data are needed to define safety, including pathogen reduction, and functionality of the obtained PL. In addition, relevant questions concerning the definition of PL release criteria, including concentration ranges of specific growth factors in PL batches for various clinical indications, also need to be addressed. We are still far from a common definition of PL and standardized PL manufacture due to our limited knowledge of the mechanisms that mediate PL-promoting cell growth. Here, we concisely discuss aspects of PL as MSC culture supplement as a preliminary step towards an agreed definition of the required characteristics of PL for the requirements of manufacturers and users.
Collapse
Affiliation(s)
- Giuseppe Astori
- Advanced Cellular Therapy Laboratory, Department of Cellular Therapy and Hematology, San Bortolo Hospital, Via Rodolfi 37, 36100, Vicenza, Italy.
| | - Eliana Amati
- Advanced Cellular Therapy Laboratory, Department of Cellular Therapy and Hematology, San Bortolo Hospital, Via Rodolfi 37, 36100, Vicenza, Italy
| | - Franco Bambi
- Transfusion Medicine and Cell Therapy, "A. Meyer" University Children's Hospital, Florence, Italy
| | - Martina Bernardi
- Advanced Cellular Therapy Laboratory, Department of Cellular Therapy and Hematology, San Bortolo Hospital, Via Rodolfi 37, 36100, Vicenza, Italy.,Hematology Project Foundation, Contrà S. Francesco 41, Vicenza, Italy
| | - Katia Chieregato
- Advanced Cellular Therapy Laboratory, Department of Cellular Therapy and Hematology, San Bortolo Hospital, Via Rodolfi 37, 36100, Vicenza, Italy.,Hematology Project Foundation, Contrà S. Francesco 41, Vicenza, Italy
| | - Richard Schäfer
- Department of Cell Therapeutics & Cell Processing, Institute for Transfusion Medicine and Immunohaematology, German Red Cross Blood Donor Service, Baden-Württemberg-Hessen gGmbH, Goethe-University Hospital, Sandhofstrasse 1, Frankfurt am Main, Germany
| | - Sabrina Sella
- Advanced Cellular Therapy Laboratory, Department of Cellular Therapy and Hematology, San Bortolo Hospital, Via Rodolfi 37, 36100, Vicenza, Italy
| | - Francesco Rodeghiero
- Advanced Cellular Therapy Laboratory, Department of Cellular Therapy and Hematology, San Bortolo Hospital, Via Rodolfi 37, 36100, Vicenza, Italy.,Hematology Project Foundation, Contrà S. Francesco 41, Vicenza, Italy
| |
Collapse
|
36
|
Kashiwazaki D, Akioka N, Kuwayama N, Hayashi T, Noguchi K, Tanaka K, Kuroda S. Involvement of circulating endothelial progenitor cells in carotid plaque growth and vulnerability. J Neurosurg 2016; 125:1549-1556. [PMID: 26871204 DOI: 10.3171/2015.10.jns151500] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The roles of endothelial progenitor cells (EPCs) in the development of carotid plaque are still obscure. This study aimed to clarify this by assessing the histological findings of specimens obtained from carotid endarterectomy. METHODS This study included 34 patients who underwent carotid endarterectomy. MR imaging was performed to semiquantitatively analyze the components of the carotid plaques in all patients. The surgical specimens were subjected to immunohistochemistry. The distributions of the CD34-, CD133-, VEGF-2R-positive cells in the carotid plaques were precisely analyzed, and their number was quantified. Simultaneously, the CD34-positive microvessels were localized. RESULTS The plaque component was judged as lipid-rich plaque in 19 patients, intraplaque hemorrhage (IPH) in 11 patients, and fibrous plaque in 4 patients. The CD34-positive microvessels were densely distributed in the plaque shoulder and interface-to-media regions. The CD34-, CD133-, and VEGF-2R-positive cells were mainly localized around the CD34-positive microvessels. The number of CD34-positive microvessels significantly correlated with the number of CD34-, CD133-, and VEGF-2R-positive cells (R = 0.308, p = 0.009; R = 0.324, p = 0.006; and R = 0.296, p = 0.013, respectively). Vulnerable plaques (lipid-rich and IPH) had significantly higher numbers of the CD34-positive microvessels (p = 0.007) and CD34-, CD133-, and VEGF-2R-positive cells than fibrous plaques (p = 0.031, p = 0.013, and p = 0.002). CONCLUSIONS These findings strongly suggest that neovascularization in the plaque shoulder and interface-to-media regions may play a key role in delivering EPCs from the peripheral blood to the carotid plaque, promoting the growth of carotid plaque. Furthermore, the invaded EPCs, especially the CD133-positive immature EPCs, may be related to plaque vulnerability.
Collapse
Affiliation(s)
| | | | | | | | - Kyo Noguchi
- Radiology, Graduate School of Medicine and Pharmacological Sciences, University of Toyama; and
| | - Kortaro Tanaka
- Department of Neurology, Toyama University Hospital, Toyama, Japan
| | | |
Collapse
|
37
|
Jin K, Li B, Lou L, Xu Y, Ye X, Yao K, Ye J, Gao C. In vivo vascularization of MSC-loaded porous hydroxyapatite constructs coated with VEGF-functionalized collagen/heparin multilayers. Sci Rep 2016; 6:19871. [PMID: 26794266 PMCID: PMC4726420 DOI: 10.1038/srep19871] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 12/18/2015] [Indexed: 12/18/2022] Open
Abstract
Rapid and adequate vascularization is vital to the long-term success of porous orbital enucleation implants. In this study, porous hydroxyapatite (HA) scaffolds coated with vascular endothelial growth factor (VEGF)-functionalized collagen (COL)/heparin (HEP) multilayers (porosity 75%, pore size 316.8 ± 77.1 μm, VEGF dose 3.39 ng/mm3) were fabricated to enhance vascularization by inducing the differentiation of mesenchymal stem cells (MSCs) to endothelial cells. The in vitro immunofluorescence staining, quantitative real-time polymerase chain reaction (qRT-PCR), and western blotting results demonstrated that the expression of the endothelial differentiation markers CD31, Flk-1, and von Willebrand factor (vWF) was significantly increased in the HA/(COL/HEP)5/VEGF/MSCs group compared with the HA/VEGF/MSCs group. Moreover, the HA/(COL/HEP)5 scaffolds showed a better entrapment of the MSCs and accelerated cell proliferation. The in vivo assays showed that the number of newly formed vessels within the constructs after 28 d was significantly higher in the HA/(COL/HEP)5/VEGF/MSCs group (51.9 ± 6.3/mm2) than in the HA (26.7 ± 2.3/mm2) and HA/VEGF/MSCs (38.2 ± 2.4/mm2) groups. The qRT-PCR and western blotting results demonstrated that the HA/(COL/HEP)5/VEGF/MSCs group also had the highest expression of CD31, Flk-1, and vWF at both the mRNA and protein levels.
Collapse
Affiliation(s)
- Kai Jin
- Department of Ophthalmology, the Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou 310009, China
| | - Bo Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Lixia Lou
- Department of Ophthalmology, the Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou 310009, China
| | - Yufeng Xu
- Department of Ophthalmology, the Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou 310009, China
| | - Xin Ye
- Department of Ophthalmology, the Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou 310009, China
| | - Ke Yao
- Department of Ophthalmology, the Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou 310009, China
| | - Juan Ye
- Department of Ophthalmology, the Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou 310009, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
38
|
Laschke MW, Menger MD. Prevascularization in tissue engineering: Current concepts and future directions. Biotechnol Adv 2015; 34:112-21. [PMID: 26674312 DOI: 10.1016/j.biotechadv.2015.12.004] [Citation(s) in RCA: 184] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/16/2015] [Accepted: 12/04/2015] [Indexed: 12/24/2022]
Abstract
The survival of engineered tissue constructs during the initial phase after their implantation depends on the rapid development of an adequate vascularization. This, in turn, is a major prerequisite for the constructs' long-term function. 'Prevascularization' has emerged as a promising concept in tissue engineering, aiming at the generation of a preformed microvasculature in tissue constructs prior to their implantation. This should shorten the time period during which the constructs are avascular and suffer hypoxic conditions. Herein, we provide an overview of current strategies for the generation of preformed microvascular networks within tissue constructs. In vitro approaches use cell seeding, spheroid formation or cell sheet technologies. In situ approaches use the body as a natural bioreactor to induce vascularization by angiogenic ingrowth or flap and arteriovenous (AV)-loop techniques. In future, these strategies may be supplemented by the transplantation of adipose tissue-derived microvascular fragments or the in vitro generation of highly organized microvascular networks by means of sophisticated microscale technologies and microfluidic systems. The further advancement of these prevascularization concepts and their adaptation to individual therapeutic interventions will markedly contribute to a broad implementation of tissue engineering applications into clinical practice.
Collapse
Affiliation(s)
- Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, D-66421 Homburg/Saar, Germany.
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, D-66421 Homburg/Saar, Germany
| |
Collapse
|
39
|
Jalowiec JM, D'Este M, Bara JJ, Denom J, Menzel U, Alini M, Verrier S, Herrmann M. An In Vitro Investigation of Platelet-Rich Plasma-Gel as a Cell and Growth Factor Delivery Vehicle for Tissue Engineering. Tissue Eng Part C Methods 2015; 22:49-58. [PMID: 26467221 DOI: 10.1089/ten.tec.2015.0223] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Platelet-rich plasma (PRP) has been used for different applications in human and veterinary medicine. Many studies have shown promising therapeutic effects of PRP; however, there are still many controversies regarding its composition, properties, and clinical efficacy. The aim of this study was to evaluate the influence of different platelet concentrations on the rheological properties and growth factor (GF) release profile of PRP-gels. In addition, the viability of incorporated bone marrow-derived human mesenchymal stem cells (MSCs) was investigated. PRP (containing 1000 × 10(3), 2000 × 10(3), and 10,000 × 10(3) platelets/μL) was prepared from human platelet concentrates. Platelet activation and gelification were achieved by addition of human thrombin. Viscoelastic properties of PRP-gels were evaluated by rheological studies. The release of GFs and inflammatory proteins was measured using a membrane-based protein array and enzyme-linked immunosorbent assay. MSC viability and proliferation in PRP-gels were assessed over 7 days by cell viability staining. Cell proliferation was examined using DNA quantification. Regardless of the platelet content, all tested PRP-gels showed effective cross-linking. A positive correlation between protein release and the platelet concentration was observed at all time points. Among the detected proteins, the chemokine CCL5 was the most abundant. The greatest release appeared within the first 4 h after gelification. MSCs could be successfully cultured in PRP-gels over 7 days, with the highest cell viability and DNA content found in PRP-gels with 1000 × 10(3) platelets/μL. The results of this study suggest that PRP-gels represent a suitable carrier for both cell and GF delivery for tissue engineering. Notably, a platelet concentration of 1000 × 10(3) platelets/μL appeared to provide the most favorable environment for MSCs. Thus, the platelet concentration is an important consideration for the clinical application of PRP-gels.
Collapse
Affiliation(s)
| | | | | | - Jessica Denom
- 2 Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Université Paris Diderot Paris 7 , Paris, France
| | | | - Mauro Alini
- 1 AO Research Institute Davos , Davos, Switzerland
| | | | | |
Collapse
|
40
|
Heme-Mediated Induction of CXCL10 and Depletion of CD34+ Progenitor Cells Is Toll-Like Receptor 4 Dependent. PLoS One 2015; 10:e0142328. [PMID: 26555697 PMCID: PMC4640861 DOI: 10.1371/journal.pone.0142328] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 10/19/2015] [Indexed: 11/19/2022] Open
Abstract
Plasmodium falciparum infection can cause microvascular dysfunction, cerebral encephalopathy and death if untreated. We have previously shown that high concentrations of free heme, and C-X-C motif chemokine 10 (CXCL10) in sera of malaria patients induce apoptosis in microvascular endothelial and neuronal cells contributing to vascular dysfunction, blood-brain barrier (BBB) damage and mortality. Endothelial progenitor cells (EPC) are microvascular endothelial cell precursors partly responsible for repair and regeneration of damaged BBB endothelium. Studies have shown that EPC's are depleted in severe malaria patients, but the mechanisms mediating this phenomenon are unknown. Toll-like receptors recognize a wide variety of pathogen-associated molecular patterns generated by pathogens such as bacteria and parasites. We tested the hypothesis that EPC depletion during malaria pathogenesis is a function of heme-induced apoptosis mediated by CXCL10 induction and toll-like receptor (TLR) activation. Heme and CXCL10 concentrations in plasma obtained from malaria patients were elevated compared with non-malaria subjects. EPC numbers were significantly decreased in malaria patients (P < 0.02) and TLR4 expression was significantly elevated in vivo. These findings were confirmed in EPC precursors in vitro; where it was determined that heme-induced apoptosis and CXCL10 expression was TLR4-mediated. We conclude that increased serum heme mediates depletion of EPC during malaria pathogenesis.
Collapse
|
41
|
Burnouf T, Strunk D, Koh MBC, Schallmoser K. Human platelet lysate: Replacing fetal bovine serum as a gold standard for human cell propagation? Biomaterials 2015; 76:371-87. [PMID: 26561934 DOI: 10.1016/j.biomaterials.2015.10.065] [Citation(s) in RCA: 357] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/16/2015] [Accepted: 10/26/2015] [Indexed: 02/07/2023]
Abstract
The essential physiological role of platelets in wound healing and tissue repair builds the rationale for the use of human platelet derivatives in regenerative medicine. Abundant growth factors and cytokines stored in platelet granules can be naturally released by thrombin activation and clotting or artificially by freeze/thaw-mediated platelet lysis, sonication or chemical treatment. Human platelet lysate prepared by the various release strategies has been established as a suitable alternative to fetal bovine serum as culture medium supplement, enabling efficient propagation of human cells under animal serum-free conditions for a multiplicity of applications in advanced somatic cell therapy and tissue engineering. The rapidly increasing number of studies using platelet derived products for inducing human cell proliferation and differentiation has also uncovered a considerable variability of human platelet lysate preparations which limits comparability of results. The main variations discussed herein encompass aspects of donor selection, preparation of the starting material, the possibility for pooling in plasma or additive solution, the implementation of pathogen inactivation and consideration of ABO blood groups, all of which can influence applicability. This review outlines the current knowledge about human platelet lysate as a powerful additive for human cell propagation and highlights its role as a prevailing supplement for human cell culture capable to replace animal serum in a growing spectrum of applications.
Collapse
Affiliation(s)
- Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Dirk Strunk
- Experimental & Clinical Cell Therapy Institute, Paracelsus Medical University, Salzburg, Austria; Spinal Cord Injury & Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Mickey B C Koh
- Blood Services Group, Health Sciences Authority, Singapore; Department for Hematology, St George's Hospital and Medical School, London, UK
| | - Katharina Schallmoser
- Spinal Cord Injury & Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria; Department for Blood Group Serology and Transfusion Medicine, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
42
|
Endothelial Progenitor Cell Fraction Contained in Bone Marrow-Derived Mesenchymal Stem Cell Populations Impairs Osteogenic Differentiation. BIOMED RESEARCH INTERNATIONAL 2015; 2015:659542. [PMID: 26491682 PMCID: PMC4600555 DOI: 10.1155/2015/659542] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 07/28/2015] [Indexed: 12/11/2022]
Abstract
In bone tissue engineering (TE) endothelial cell-osteoblast cocultures are known to induce synergies of cell differentiation and activity. Bone marrow mononucleated cells (BMCs) are a rich source of mesenchymal stem cells (MSCs) able to develop an osteogenic phenotype. Endothelial progenitor cells (EPCs) are also present within BMC. In this study we investigate the effect of EPCs present in the BMC population on MSCs osteogenic differentiation. Human BMCs were isolated and separated into two populations. The MSC population was selected through plastic adhesion capacity. EPCs (CD34+ and CD133+) were removed from the BMC population and the resulting population was named depleted MSCs. Both populations were cultured over 28 days in osteogenic medium (Dex+) or medium containing platelet lysate (PL). MSC population grew faster than depleted MSCs in both media, and PL containing medium accelerated the proliferation for both populations. Cell differentiation was much higher in Dex+ medium in both cases. Real-time RT-PCR revealed upregulation of osteogenic marker genes in depleted MSCs. Higher values of ALP activity and matrix mineralization analyses confirmed these results. Our study advocates that absence of EPCs in the MSC population enables higher osteogenic gene expression and matrix mineralization and therefore may lead to advanced bone neoformation necessary for TE constructs.
Collapse
|