1
|
Shen Y, Nakajima H, Zhu J, Wu W. Integrin β2 regulates titanium particle‑induced inflammation in macrophages: In vitro aseptic loosening model. Mol Med Rep 2025; 31:25. [PMID: 39540364 DOI: 10.3892/mmr.2024.13390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024] Open
Abstract
Aseptic loosening is a major complication of joint replacement surgery, characterized by periprosthetic osteolysis and chronic inflammation at the bone‑implant interface. Cells release chemokines, cytokines and other pro‑inflammatory substances that perpetuate inflammation reactions, while other particle‑stimulated macrophages promote osteoclastic bone resorption and impair bone formation. The present study investigated integrin and inflammatory cytokine expression patterns in RAW 264.7 cells treated with titanium (Ti) particles to elucidate the role of integrins in Ti particle‑mediated inflammatory osteolysis. Assessment was performed by reverse transcription‑quantitative PCR, western blotting, confocal immunofluorescence, flow cytometry and enzyme‑linked immunosorbent assays. Cell migration was evaluated by wound healing assay. It was found that Ti particles significantly induced integrin expression in RAW 264.7 cells, including upregulation of integrins β2 (CD18), aL (CD11a), aM (CD11b) and aX (CD11c). Ti particles also enhanced the expression of Toll‑like receptors (TLRs; TLR1, TLR2, TLR3 and TLR4) and triggered the release of inflammatory cytokines such as tumor necrosis factor α, interleukin (IL)‑1β, IL‑8 and IL‑12. Proteomics showed higher expression and activity levels of TLR2 and TLR4, along with their downstream signaling adaptors myeloid differentiation primary response protein 88 (MyD88) and Mal/TIR‑domain‑containing adapter protein (TIRAP), following Ti treatment. Additionally, Ti treatment significantly enhanced the migration rate of RAW 264.7 cells. The present findings indicated that Ti particles regulate the inflammatory response of RAW 264.7 cells in an in vitro aseptic loosening model by activating the TLR/TIRAP/MyD88 signaling pathway.
Collapse
Affiliation(s)
- Yue Shen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Haruna Nakajima
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113‑8654, Japan
| | - Junfeng Zhu
- Department of Orthopedics Surgery, Suichang Branch of The Second Affiliated Hospital, Zhejiang University School of Medicine (Suichang County People's Hospital in Zhejiang), Lishui, Zhejiang 323300, P.R. China
| | - Weigang Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| |
Collapse
|
2
|
Leung CK, Zhu P, Loke I, Tang KF, Leung HC, Yeung CF. Development of a quantitative prediction algorithm for human cord blood-derived CD34 + hematopoietic stem-progenitor cells using parametric and non-parametric machine learning models. Sci Rep 2024; 14:25085. [PMID: 39443591 PMCID: PMC11500098 DOI: 10.1038/s41598-024-75731-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
The transplantation of CD34+ hematopoietic stem-progenitor cells (HSPCs) derived from cord blood serves as the standard treatment for selected hematological, oncological, metabolic, and immunodeficiency disorders, of which the dose is pivotal to the clinical outcome. Based on numerous maternal and neonatal parameters, we evaluated the predictive power of mathematical pipelines to the proportion of CD34+ cells in the final cryopreserved cord blood product adopting both parametric and non-parametric algorithms. Twenty-four predictor variables associated with the cord blood processing of 802 processed cord blood units randomly sampled in 2020-2022 were retrieved and analyzed. Prediction models were developed by adopting the parametric (multivariate linear regression) and non-parametric (random forest and back propagation neural network) statistical models to investigate the data patterns for determining the single outcome (i.e., the proportion of CD34+ cells). The multivariate linear regression model produced the lowest root-mean-square deviation (0.0982). However, the model created by the back propagation neural network produced the highest median absolute deviation (0.0689) and predictive power (56.99%) in comparison to the random forest and multivariate linear regression. The predictive model depending on a combination of continuous and discrete maternal with neonatal parameters associated with cord blood processing can predict the CD34+ dose in the final product for clinical utilization. The back propagation neural network algorithm produces a model with the highest predictive power which can be widely applied to assisting cell banks for optimal cord blood unit selection to ensure the highest chance of transplantation success.
Collapse
Affiliation(s)
- Chi-Kwan Leung
- Group Laboratory Operations, Cordlife Group Limited, A'Posh Bizhub #06-01/09, 1 Yishun Industrial Street 1, Singapore, 768160, Singapore.
| | - Pengcheng Zhu
- Group Laboratory Operations, Cordlife Group Limited, A'Posh Bizhub #06-01/09, 1 Yishun Industrial Street 1, Singapore, 768160, Singapore
| | - Ian Loke
- Group Laboratory Operations, Cordlife Group Limited, A'Posh Bizhub #06-01/09, 1 Yishun Industrial Street 1, Singapore, 768160, Singapore
| | - Kin Fai Tang
- Group Laboratory Operations, Cordlife Group Limited, A'Posh Bizhub #06-01/09, 1 Yishun Industrial Street 1, Singapore, 768160, Singapore
| | - Ho-Chuen Leung
- Group Laboratory Operations, Cordlife Group Limited, A'Posh Bizhub #06-01/09, 1 Yishun Industrial Street 1, Singapore, 768160, Singapore
| | - Chin-Fung Yeung
- Group Laboratory Operations, Cordlife Group Limited, A'Posh Bizhub #06-01/09, 1 Yishun Industrial Street 1, Singapore, 768160, Singapore
| |
Collapse
|
3
|
El-Serafi I, Steele S. Cyclophosphamide Pharmacogenomic Variation in Cancer Treatment and Its Effect on Bioactivation and Pharmacokinetics. Adv Pharmacol Pharm Sci 2024; 2024:4862706. [PMID: 38966316 PMCID: PMC11223907 DOI: 10.1155/2024/4862706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 07/06/2024] Open
Abstract
Cyclophosphamide (Cy) is a prodrug that is mainly bioactivated by cytochrome P450 (CYP) 2B6 enzyme. Several other enzymes are also involved in its bioactivation and affect its kinetics. Previous studies have shown the effect of the enzymes' genetic polymorphisms on Cy kinetics and its clinical outcome. These results were controversial primarily because of the involvement of several interacting enzymes in the Cy metabolic pathway, which can also be affected by several clinical factors as well as other drug interactions. In this review article, we present the effect of CYP2B6 polymorphisms on Cy kinetics since it is the main bioactivating enzyme, as well as discussing all previously reported enzymes and clinical factors that can alter Cy efficacy. Additionally, we present explanations for key Cy side effects related to the nature and site of its bioactivation. Finally, we discuss the role of busulphan in conditioning regimens in the Cy metabolic pathway as a clinical example of drug-drug interactions involving several enzymes. By the end of this article, our aim is to have provided a comprehensive summary of Cy pharmacogenomics and the effect on its kinetics. The utility of these findings in the development of new strategies for Cy personalized patient dose adjustment will aid in the future optimization of patient specific Cy dosages and ultimately in improving clinical outcomes. In conclusion, CYP2B6 and several other enzyme polymorphisms can alter Cy kinetics and consequently the clinical outcomes. However, the precise quantification of Cy kinetics in any individual patient is complex as it is clearly under multifactorial genetic control. Additionally, other clinical factors such as the patient's age, diagnosis, concomitant medications, and clinical status should also be considered.
Collapse
Affiliation(s)
- Ibrahim El-Serafi
- Basic Medical Sciences DepartmentCollege of MedicineAjman University, Ajman, UAE
- Department of Hand Surgery, and Plastic Surgery and BurnsLinköping University Hospital, Linkoöping, Sweden
| | - Sinclair Steele
- Pathological Sciences DepartmentCollege of MedicineAjman University, Ajman, UAE
| |
Collapse
|
4
|
Liu Y, Yu S, Chen Y, Hu Z, Fan L, Liang G. The clinical regimens and cell membrane camouflaged nanodrug delivery systems in hematologic malignancies treatment. Front Pharmacol 2024; 15:1376955. [PMID: 38689664 PMCID: PMC11059051 DOI: 10.3389/fphar.2024.1376955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/02/2024] [Indexed: 05/02/2024] Open
Abstract
Hematologic malignancies (HMs), also referred to as hematological or blood cancers, pose significant threats to patients as they impact the blood, bone marrow, and lymphatic system. Despite significant clinical strategies using chemotherapy, radiotherapy, stem cell transplantation, targeted molecular therapy, or immunotherapy, the five-year overall survival of patients with HMs is still low. Fortunately, recent studies demonstrate that the nanodrug delivery system holds the potential to address these challenges and foster effective anti-HMs with precise treatment. In particular, cell membrane camouflaged nanodrug offers enhanced drug targeting, reduced toxicity and side effects, and/or improved immune response to HMs. This review firstly introduces the merits and demerits of clinical strategies in HMs treatment, and then summarizes the types, advantages, and disadvantages of current nanocarriers helping drug delivery in HMs treatment. Furthermore, the types, functions, and mechanisms of cell membrane fragments that help nanodrugs specifically targeted to and accumulate in HM lesions are introduced in detail. Finally, suggestions are given about their clinical translation and future designs on the surface of nanodrugs with multiple functions to improve therapeutic efficiency for cancers.
Collapse
Affiliation(s)
- Yuanyuan Liu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Shanwu Yu
- College of Horticulture and Plant Protection, Henan University of Science and Technology, Luoyang, Henan, China
| | - Yixiang Chen
- Luoyang Vocational and Technical College, Luoyang, Henan, China
| | - Zhihong Hu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Lingling Fan
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Gaofeng Liang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| |
Collapse
|
5
|
Blitzer GC, Paz C, Glassey A, Ganz OR, Giri J, Pennati A, Meyers RO, Bates AM, Nickel KP, Weiss M, Morris ZS, Mattison RJ, McDowell KA, Croxford E, Chappell RJ, Glazer TA, Rogus-Pulia NM, Galipeau J, Kimple RJ. Functionality of bone marrow mesenchymal stromal cells derived from head and neck cancer patients - A FDA-IND enabling study regarding MSC-based treatments for radiation-induced xerostomia. Radiother Oncol 2024; 192:110093. [PMID: 38224919 PMCID: PMC10922976 DOI: 10.1016/j.radonc.2024.110093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/17/2024]
Abstract
PURPOSE Salivary dysfunction is a significant side effect of radiation therapy for head and neck cancer (HNC). Preliminary data suggests that mesenchymal stromal cells (MSCs) can improve salivary function. Whether MSCs from HNC patients who have completed chemoradiation are functionally similar to those from healthy patients is unknown. We performed a pilot clinical study to determine whether bone marrow-derived MSCs [MSC(M)] from HNC patients could be used for the treatment of RT-induced salivary dysfunction. METHODS An IRB-approved pilot clinical study was undertaken on HNC patients with xerostomia who had completed treatment two or more years prior. Patients underwent iliac crest bone marrow aspirate and MSC(M) were isolated and cultured. Culture-expanded MSC(M) were stimulated with IFNγ and cryopreserved prior to reanimation and profiling for functional markers by flow cytometry and ELISA. MSC(M) were additionally injected into mice with radiation-induced xerostomia and the changes in salivary gland histology and salivary production were examined. RESULTS A total of six subjects were enrolled. MSC(M) from all subjects were culture expanded to > 20 million cells in a median of 15.5 days (range 8-20 days). Flow cytometry confirmed that cultured cells from HNC patients were MSC(M). Functional flow cytometry demonstrated that these IFNγ-stimulated MSC(M) acquired an immunosuppressive phenotype. IFNγ-stimulated MSC(M) from HNC patients were found to express GDNF, WNT1, and R-spondin 1 as well as pro-angiogenesis and immunomodulatory cytokines. In mice, IFNγ-stimulated MSC(M) injection after radiation decreased the loss of acinar cells, decreased the formation of fibrosis, and increased salivary production. CONCLUSIONS MSC (M) from previously treated HNC patients can be expanded for auto-transplantation and are functionally active. Furthermore IFNγ-stimulated MSC(M) express proteins implicated in salivary gland regeneration. This study provides preliminary data supporting the feasibility of using autologous MSC(M) from HNC patients to treat RT-induced salivary dysfunction.
Collapse
Affiliation(s)
- Grace C Blitzer
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Cristina Paz
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Annemarie Glassey
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Olga R Ganz
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Jayeeta Giri
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Andrea Pennati
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; UW Carbone Cancer Center, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Ross O Meyers
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Amber M Bates
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Kwangok P Nickel
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Marissa Weiss
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Zachary S Morris
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Ryan J Mattison
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; UW Carbone Cancer Center, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Kimberly A McDowell
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Emma Croxford
- Department of Biostatistics and Medical Informatics, 610 Walnut Street, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53726 USA
| | - Richard J Chappell
- Department of Biostatistics and Medical Informatics, 610 Walnut Street, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53726 USA; UW Carbone Cancer Center, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Tiffany A Glazer
- Department of Surgery, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Nicole M Rogus-Pulia
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; UW Carbone Cancer Center, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; Geriatric Research Education and Clinical Center, 2500 Overlook Terrace, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Jacques Galipeau
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; UW Carbone Cancer Center, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Randall J Kimple
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; UW Carbone Cancer Center, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA.
| |
Collapse
|
6
|
Khaled SAA, Elzembely MM, Soliman AMA, Shwakat N, Rafaat N, Malek MA, Abdelmageed ES. Effective and Elaborative Induction Program for Mitigating Myths and Misconceptions Linked to Hematopoietic Stem Cell Transplantation in a Resource Limited Setting. Indian J Hematol Blood Transfus 2023; 39:598-609. [PMID: 37786824 PMCID: PMC10542043 DOI: 10.1007/s12288-023-01634-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 02/07/2023] [Indexed: 03/29/2023] Open
Abstract
Since the first transplant in 1957 and hematopoietic stem cell transplantation (HSCT) is the curative modality for numerous hematological disorders. Nevertheless, it is not available for all patients. Besides unavailability of matched donors a lot of factors could hinder HSCT in a resource limited setting, as financial and administrative factors. In our daily practice we noticed other factors that hinder HSCT in our center, the common myths and misconceptions about HSCT and donation. This quasi-experimental study assessed, for the first time, common myths and misconceptions about HSCT among 218 medical and nursing students before and after an interventional educational program. The study tool was an investigators' developed self-administered questionnaire. Participants' male to female ratio was 1:2.5, and FAS was middle in 52.7%. Pretest high myths scores were reported in 53.4% and 90% of medical and nursing students that was reduced to 0% and 4% post-test, respectively. Pretest, 26.3% and 7% of medical and nursing students welling to donate HSC, that increased to 66% and 39% post-test, respectively. Rural residency, low and middle FAS associated with higher myths scores. Myths score is an independent effector of willingness to donate HSC among participants. In conclusion medical/nursing students had significant myths and misconceptions about HSCT that was corrected with the educational program. Thus, wide based educational programs about HSCT are mandatory to correct myths and augment HSC donation. www.clinicaltrrial.gov: clinical trial ID NCT05151406. Supplementary Information The online version contains supplementary material available at 10.1007/s12288-023-01634-5.
Collapse
Affiliation(s)
- Safaa A. A. Khaled
- Department of Internal Medicine-Clinical Hematology Unit, Assiut University Hospital, Assiut, Egypt
- Unit of Bone Marrow Transplantation, South Egypt Cancer Institute, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Mahmoud M. Elzembely
- Department of Pediatric Oncology, South Egypt Cancer Institute, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Asmaa M. A. Soliman
- Department of Public Health and Community Medicine, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Nahed Shwakat
- Department of Nursing Administration, Faculty of Nursing, Assiut University, Assiut, Egypt
| | - Nashwa Rafaat
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | | | - Esmat S. Abdelmageed
- Department of Medical Surgical Nursing, Faculty of Nursing, Assiut University, Assiut, Egypt
| |
Collapse
|
7
|
Jiang X, Li W, Ge L, Lu M. Mesenchymal Stem Cell Senescence during Aging:From Mechanisms to Rejuvenation Strategies. Aging Dis 2023; 14:1651-1676. [PMID: 37196126 PMCID: PMC10529739 DOI: 10.14336/ad.2023.0208] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/08/2023] [Indexed: 05/19/2023] Open
Abstract
In cell transplantation therapy, mesenchymal stem cells(MSCs)are ideal seed cells due to their easy acquisition and cultivation, strong regenerative capacity, multi-directional differentiation abilities, and immunomodulatory effects. Autologous MSCs are better applicable compared with allogeneic MSCs in clinical practice. The elderly are the main population for cell transplantation therapy, but as donor aging, MSCs in the tissue show aging-related changes. When the number of generations of in vitro expansion is increased, MSCs will also exhibit replicative senescence. The quantity and quality of MSCs decline during aging, which limits the efficacy of autologous MSCs transplantation therapy. In this review, we examine the changes in MSC senescence as a result of aging, discuss the progress of research on mechanisms and signalling pathways of MSC senescence, and discuss possible rejuvenation strategies of aged MSCs to combat senescence and enhance the health and therapeutic potential of MSCs.
Collapse
Affiliation(s)
- Xinchen Jiang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China.
- Hunan provincical key laboratory of Neurorestoratology, the Second Affiliated Hospital, Hunan Normal University, Changsha, China.
| | - Wenshui Li
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China.
- Hunan provincical key laboratory of Neurorestoratology, the Second Affiliated Hospital, Hunan Normal University, Changsha, China.
| | - Lite Ge
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China.
- Hunan provincical key laboratory of Neurorestoratology, the Second Affiliated Hospital, Hunan Normal University, Changsha, China.
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China, Changsha
| | - Ming Lu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China.
- Hunan provincical key laboratory of Neurorestoratology, the Second Affiliated Hospital, Hunan Normal University, Changsha, China.
| |
Collapse
|
8
|
Al-Mutheffer EA, Reinwald Y, El Haj AJ. Donor variability of ovine bone marrow derived mesenchymal stem cell - implications for cell therapy. Int J Vet Sci Med 2023; 11:23-37. [PMID: 37092030 PMCID: PMC10114964 DOI: 10.1080/23144599.2023.2197393] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 02/20/2023] [Indexed: 04/25/2023] Open
Abstract
It is assumed that all species, including sheep, demonstrate significant variation between individuals including the characteristics of their bone marrow-derived mesenchymal stem cells (BM-MSCs). These differences may account for limited success in pre-clinical animal studies and may also impact on treatment strategies that are used within regenerative medicine. This study investigates variations between ovine MSCs (oMSCs) isolated from 13 English Mule sheep donors by studying cell viability, expansion, the cells' trilineage differentiation potential and the expression of cell surface markers. In addition to the primary objective, this article also compares various differentiation media used for the trilineage differentiation of oMSCs. In this study, a clear individual variation between the sheep donors regarding oMSCs characterization, tri-lineage differentiation potential and marker expression was effectively demonstrated. The results set out to systematically explore the ovine mesenchymal stem cell population derived from multiple donors. With this information, it is possible to start addressing the issues of personalized approaches to regenerative therapies.
Collapse
Affiliation(s)
- E’atelaf A. Al-Mutheffer
- Institute for Science and Technology in Medicine, Keele University, Stoke-on-Trent, UK
- Department of Surgery and Obstetrics, College of Veterinary Medicine, Baghdad University, Baghdad, Iraq
| | - Yvonne Reinwald
- Institute for Science and Technology in Medicine, Keele University, Stoke-on-Trent, UK
- School of Science and Technology, Department of Engineering, Nottingham Trent University Nottingham, Nottingham, UK
| | - Alicia J. El Haj
- Institute for Science and Technology in Medicine, Keele University, Stoke-on-Trent, UK
- School of Chemical Engineering, Healthcare Technology Institute, Institute of Translational Medicine Birmingham University, Birmingham, UK
| |
Collapse
|
9
|
Motta CM, Keller MD, Bollard CM. Applications of Virus specific T cell Therapies Post BMT. Semin Hematol 2022; 60:10-19. [PMID: 37080705 DOI: 10.1053/j.seminhematol.2022.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) has been used as a curative standard of care for moderate to severe primary immunodeficiency disorders as well as relapsed hematologic malignancies for over 50 years [1,2]. However, chronic and refractory viral infections remain a leading cause of morbidity and mortality in the immune deficient period following HSCT, where use of available antiviral pharmacotherapies is limited by toxicity and emerging resistance [3]. Adoptive immunotherapy using virus-specific T cells (VSTs) has been explored for over 2 decades [4,5] in patients post-HSCT and has been shown prior phase I-II studies to be safe and effective for treatment or preventions of viral infections including cytomegalovirus, Epstein-Barr virus, BK virus, and adenovirus with minimal toxicity and low risk of graft vs host disease [6-9]. This review summarizes methodologies to generate VSTs the clinical results utilizing VST therapeutics and the challenges and future directions for the field.
Collapse
|
10
|
Blitzer GC, Rogus-Pulia NM, Mattison RJ, Varghese T, Ganz O, Chappell R, Galipeau J, McDowell KA, Meyers RO, Glazer TA, Kimple RJ. Marrow-Derived Autologous Stromal Cells for the Restoration of Salivary Hypofunction (MARSH): Study protocol for a phase 1 dose-escalation trial of patients with xerostomia after radiation therapy for head and neck cancer: MARSH: Marrow-Derived Autologous Stromal Cells for the Restoration of Salivary Hypofunction. Cytotherapy 2022; 24:534-543. [PMID: 35183442 PMCID: PMC9038658 DOI: 10.1016/j.jcyt.2021.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/11/2021] [Accepted: 11/03/2021] [Indexed: 01/28/2023]
Abstract
BACKGROUND Xerostomia, or dry mouth, is a common side effect of head and neck radiation. Current treatment options for radiation-induced xerostomia are generally supportive in nature. Adult stem cells are the ultimate source for replenishment of salivary gland tissue. Bone marrow-derived mesenchymal stromal cells (BM-MSCs) are a viable cell-based therapy for xerostomia. We have undertaken studies enabling U.S. Food and Drug Administration Investigational New Drug status, demonstrating the normal phenotype, intact functionality, and pro-growth secretome of interferon-γ (IFNγ)-stimulated BM-MSCs taken from patients with head and neck cancer who have undergone radiation ± chemotherapy. Here we present the protocol of MARSH, a first-in-human clinical trial of bone marrow-derived, IFNγ-activated BM-MSCs for the treatment of radiation-induced xerostomia. METHODS This single-center phase 1 dose-escalation with expansion cohort, non-placebo-controlled study will assess the safety and tolerability of BM-MSCs for the treatment of radiation-induced xerostomia in patients who had head and neck cancer. The phase 1 dose-escalation study will be a 3 + 3 design with staggered enrollment. A total of 21 to 30 subjects (9 to 18 in phase 1 study, 12 in expansion cohort) will be enrolled. The primary endpoint is determining the recommended phase 2 dose (RP2D) of IFNγ-stimulated BM-MSCs to enable further studies on the efficacy of BM-MSCs. Patients' bone marrow will be aspirated, and BM-MSCs will be expanded, stimulated with IFNγ, and injected into the submandibular gland. The RP2D will be determined by dose-limiting toxicities occurring within 1 month of BM-MSC injection. Secondary outcomes of saliva amounts and composition, ultrasound of salivary glands, and quality of life surveys will be taken at 3-, 6-, 12-, and 24-month visits. DISCUSSION Autotransplantation of IFNγ-stimulated BM-MSCs in salivary glands after radiation therapy or chemoradiation therapy may provide an innovative remedy to treat xerostomia and restore quality of life. This is the first therapy for radiation-induced xerostomia that may be curative. TRIAL REGISTRATION World Health Organization International Clinical Trials Registry Platform: NCT04489732.
Collapse
Affiliation(s)
- Grace C Blitzer
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Nicole M Rogus-Pulia
- Departments of Medicine and Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Ryan J Mattison
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Tomy Varghese
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Olga Ganz
- Program for Advanced Cell Therapy, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Richard Chappell
- Departments of Biostatistics & Medical Informatics and Statistics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Jacques Galipeau
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Kimberly A McDowell
- Program for Advanced Cell Therapy, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Ross O Meyers
- Program for Advanced Cell Therapy, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Tiffany A Glazer
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Randall J Kimple
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.
| |
Collapse
|
11
|
Kim J, Eom MR, Ji Jeong E, Choi JS, Kwon SK. Multiple stimulation with spheroids comprising salivary gland and adipose-derived stem cells enhances regeneration of radiation-damaged salivary glands. J IND ENG CHEM 2022. [DOI: 10.1016/j.jiec.2021.12.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
12
|
Silpa L, Sim R, Russell AJ. Recent Advances in Small Molecule Stimulation of Regeneration and Repair. Bioorg Med Chem Lett 2022; 61:128601. [PMID: 35123003 DOI: 10.1016/j.bmcl.2022.128601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 11/02/2022]
Abstract
Therapeutic approaches to stimulate regeneration and repair have the potential to transform healthcare and improve outcomes for patients suffering from numerous chronic degenerative diseases. To date most approaches have involved the transplantation of therapeutic cells, and while there have been a small number of clinical approvals, major hurdles exist to the routine adoption of such therapies. In recent years humans and other mammals have been shown to possess a regenerative capacity across multiple tissues and organs, and an innate regenerative and repair response has been shown to be activated in these organs in response to injury. These realisations have inspired a transformative approach in regenerative medicine: the development of new agents to directly target these innate regeneration and repair pathways. In this article we will review the current state of the art in the discovery of small molecule modulators of regeneration and their translation towards therapeutic agents, focussing specifically on the areas of neuroregeneration and cardiac regeneration.
Collapse
Affiliation(s)
- Laurence Silpa
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford OX1 3TA
| | - Rachel Sim
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford OX1 3TA
| | - Angela J Russell
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford OX1 3TA; Department of Pharmacology, University of Oxford, University of Oxford OX1 3QT.
| |
Collapse
|
13
|
Rahman MM, Islam MR, Islam MT, Harun-Or-Rashid M, Islam M, Abdullah S, Uddin MB, Das S, Rahaman MS, Ahmed M, Alhumaydhi FA, Emran TB, Mohamed AAR, Faruque MRI, Khandaker MU, Mostafa-Hedeab G. Stem Cell Transplantation Therapy and Neurological Disorders: Current Status and Future Perspectives. BIOLOGY 2022; 11:147. [PMID: 35053145 PMCID: PMC8772847 DOI: 10.3390/biology11010147] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/26/2021] [Accepted: 12/29/2021] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases are a global health issue with inadequate therapeutic options and an inability to restore the damaged nervous system. With advances in technology, health scientists continue to identify new approaches to the treatment of neurodegenerative diseases. Lost or injured neurons and glial cells can lead to the development of several neurological diseases, including Parkinson's disease, stroke, and multiple sclerosis. In recent years, neurons and glial cells have successfully been generated from stem cells in the laboratory utilizing cell culture technologies, fueling efforts to develop stem cell-based transplantation therapies for human patients. When a stem cell divides, each new cell has the potential to either remain a stem cell or differentiate into a germ cell with specialized characteristics, such as muscle cells, red blood cells, or brain cells. Although several obstacles remain before stem cells can be used for clinical applications, including some potential disadvantages that must be overcome, this cellular development represents a potential pathway through which patients may eventually achieve the ability to live more normal lives. In this review, we summarize the stem cell-based therapies that have been explored for various neurological disorders, discuss the potential advantages and drawbacks of these therapies, and examine future directions for this field.
Collapse
Affiliation(s)
- Mohammad Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.M.R.); (M.R.I.); (M.T.I.); (M.H.-O.-R.); (M.I.); (M.B.U.); (S.D.); (M.S.R.); (M.A.)
| | - Mohammad Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.M.R.); (M.R.I.); (M.T.I.); (M.H.-O.-R.); (M.I.); (M.B.U.); (S.D.); (M.S.R.); (M.A.)
| | - Mohammad Touhidul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.M.R.); (M.R.I.); (M.T.I.); (M.H.-O.-R.); (M.I.); (M.B.U.); (S.D.); (M.S.R.); (M.A.)
| | - Mohammad Harun-Or-Rashid
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.M.R.); (M.R.I.); (M.T.I.); (M.H.-O.-R.); (M.I.); (M.B.U.); (S.D.); (M.S.R.); (M.A.)
| | - Mahfuzul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.M.R.); (M.R.I.); (M.T.I.); (M.H.-O.-R.); (M.I.); (M.B.U.); (S.D.); (M.S.R.); (M.A.)
| | - Sabirin Abdullah
- Space Science Center, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia;
| | - Mohammad Borhan Uddin
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.M.R.); (M.R.I.); (M.T.I.); (M.H.-O.-R.); (M.I.); (M.B.U.); (S.D.); (M.S.R.); (M.A.)
| | - Sumit Das
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.M.R.); (M.R.I.); (M.T.I.); (M.H.-O.-R.); (M.I.); (M.B.U.); (S.D.); (M.S.R.); (M.A.)
| | - Mohammad Saidur Rahaman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.M.R.); (M.R.I.); (M.T.I.); (M.H.-O.-R.); (M.I.); (M.B.U.); (S.D.); (M.S.R.); (M.A.)
| | - Muniruddin Ahmed
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.M.R.); (M.R.I.); (M.T.I.); (M.H.-O.-R.); (M.I.); (M.B.U.); (S.D.); (M.S.R.); (M.A.)
| | - Fahad A. Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 52571, Saudi Arabia;
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | | | | | - Mayeen Uddin Khandaker
- Centre for Applied Physics and Radiation Technologies, School of Engineering and Technology, Sunway University, Bandar Sunway 47500, Selangor, Malaysia;
| | - Gomaa Mostafa-Hedeab
- Pharmacology Department & Health Sciences Research Unit, Medical College, Jouf University, Sakaka 72446, Saudi Arabia;
- Pharmacology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef 62521, Egypt
| |
Collapse
|
14
|
El-Serafi A, He R, Zheng W, Benkossou F, Oerther S, Zhao Y, Mellgren K, Gustafsson B, Heilmann C, Kanerva J, Lotfi K, Toporski J, Sundin M, Höglund M, Mattsson J, El-Serafi I, Hassan M. Vitamin D levels and busulphan kinetics in patients undergoing hematopoietic stem cell transplantation, a multicenter study. Bone Marrow Transplant 2021; 56:807-817. [PMID: 33087877 DOI: 10.1038/s41409-020-01091-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/24/2020] [Accepted: 10/08/2020] [Indexed: 11/09/2022]
Abstract
Vitamin D (Vit-D), an essential nutrient, interacts with different drugs including chemotherapeutic agents like busulphan, an alkylating agent used for conditioning prior to stem cell transplantation. The correlation between Vit-D plasma levels and busulphan clearance was investigated in an uncontrolled prospective study in patients and mice. Plasma 25(OH)D levels were measured and busulphan pharmacokinetics calculated in 81 patients. Adults received oral busulphan (n = 34) while children received busulphan orally (n = 19) or intravenously (n = 28). Patients received no Vit-D supplementation. To confirm our findings, pharmacokinetics after a single dose of busulphan (oral or intravenous) were evaluated in two groups of mice (n = 60) receiving high or standard-level Vit-D supplementation. Both busulphan clearance (P < 0.0001) and 25(OH)D levels (P = 0.0004) were significantly higher in adults compared to children. A significant negative correlation (P = 0.041) was found between busulphan clearance and 25(OH)D levels in children treated orally. No such correlation was observed in adults or in children receiving intravenous busulphan. In addition, no significant effect of Vit-D levels on busulphan pharmacokinetics in mice regardless of the administration route. In conclusion, 25(OH)D can affect oral busulphan pharmacokinetics in children and its level should be considered when personalizing oral busulphan treatment. Further studies are warranted to confirm the underlying mechanisms.
Collapse
Affiliation(s)
- Ahmed El-Serafi
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Rui He
- Experimental Cancer Medicine, Division of Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Research Center and Center of Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Wenyi Zheng
- Experimental Cancer Medicine, Division of Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Research Center and Center of Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Fadwa Benkossou
- Experimental Cancer Medicine, Division of Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Research Center and Center of Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Sandra Oerther
- Experimental Cancer Medicine, Division of Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Pre-clinical Laboratory, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Ying Zhao
- Experimental Cancer Medicine, Division of Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Research Center and Center of Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Karin Mellgren
- Department of Pediatric Oncology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Britt Gustafsson
- Department of Clinical Science, Intervention and Technology, CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Carsten Heilmann
- Pediatric Clinic, Rigshospitalet, National University Hospital, Copenhagen, Denmark
| | - Jukka Kanerva
- HUS Helsinki University Hospital and University of Helsinki, New Children's Hospital, Division of Hematology-Oncology and Stem Cell Transplantation, Helsinki, Finland
| | - Kourosh Lotfi
- Clinical Pharmacology, Division of Drug Research, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
- Department of Hematology, Linköping University Hospital, Linköping, Sweden
| | - Jacek Toporski
- Department of Pediatrics, Skåne University Hospital, Lund, Sweden
| | - Mikael Sundin
- Division of Pediatrics, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
- Section of Pediatric Hematology, Immunology and HCT, Astrid Lindgren Children's Hospital, Stockholm, Sweden
| | - Martin Höglund
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Jonas Mattsson
- Gloria and Seymour Epstein Chair in Cell Therapy and Transplantation, University of Toronto, Toronto, Canada
- Princess Margaret Cancer Centre, Toronto, ON, Canada
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Ibrahim El-Serafi
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Biochemistry, Faculty of Medicine, Port-Said University, Port-Said, Egypt
| | - Moustapha Hassan
- Experimental Cancer Medicine, Division of Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.
- Clinical Research Center and Center of Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|
15
|
Shreyash N, Sonker M, Bajpai S, Tiwary SK. Review of the Mechanism of Nanocarriers and Technological Developments in the Field of Nanoparticles for Applications in Cancer Theragnostics. ACS APPLIED BIO MATERIALS 2021; 4:2307-2334. [PMID: 35014353 DOI: 10.1021/acsabm.1c00020] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cancer cannot be controlled by the usage of drugs alone, and thus, nanotechnology is an important technique that can provide the drug with an impetus to act more effectively. There is adequate availability of anticancer drugs that are classified as alkylating agents, hormones, or antimetabolites. Nanoparticle (NP) carriers increase the residence time of the drug, thereby enhancing the survival rate of the drug, which otherwise gets washed off owing to the small size of the drug particles by the excretory system. For example, for enhancing the circulation, a coating of nonfouling polymers like PEG and dextran is done. Famous drugs such as doxorubicin (DOX) are commonly encapsulated inside the nanocomposite. The various classes of nanoparticles are used to enhance drug delivery by aiding it to fight against the tumor. Targeted therapy aims to attack the cells with features common to the cancer cells while minimizing damage to the normal cell, and these therapies work in one in four ways. Some block the cancer cells from reproducing newer cells, others release toxic substances to kill the cancer cells, some stimulate the immune system to destroy the cancer cells, and some block the growth of more blood vessels around cancer cells, which starve the cells of the nutrients, which is needed for their growth. This review aims to testify the advancements nanotechnology has brought in cancer therapy, and its statements are supported with recent research findings and clinical trial results.
Collapse
|
16
|
Shang F, Yu Y, Liu S, Ming L, Zhang Y, Zhou Z, Zhao J, Jin Y. Advancing application of mesenchymal stem cell-based bone tissue regeneration. Bioact Mater 2021; 6:666-683. [PMID: 33005830 PMCID: PMC7509590 DOI: 10.1016/j.bioactmat.2020.08.014] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 08/07/2020] [Accepted: 08/15/2020] [Indexed: 12/11/2022] Open
Abstract
Reconstruction of bone defects, especially the critical-sized defects, with mechanical integrity to the skeleton is important for a patient's rehabilitation, however, it still remains challenge. Utilizing biomaterials of human origin bone tissue for therapeutic purposes has provided a facilitated approach that closely mimics the critical aspects of natural bone tissue with regard to its properties. However, not only efficacious and safe but also cost-effective and convenient are important for regenerative biomaterials to achieve clinical translation and commercial success. Advances in our understanding of regenerative biomaterials and their roles in new bone formation potentially opened a new frontier in the fast-growing field of regenerative medicine. Taking inspiration from the role and multicomponent construction of native extracellular matrix (ECM) for cell accommodation, the ECM-mimicking biomaterials and the naturally decellularized ECM scaffolds were used to create new tissues for bone restoration. On the other hand, with the going deep in understanding of mesenchymal stem cells (MSCs), they have shown great promise to jumpstart and facilitate bone healing even in diseased microenvironments with pharmacology-based endogenous MSCs rescue/mobilization, systemic/local infusion of MSCs for cytotherapy, biomaterials-based approaches, cell-sheets/-aggregates technology and usage of subcellular vesicles of MSCs to achieve scaffolds-free or cell-free delivery system, all of them have been shown can improve MSCs-mediated regeneration in preclinical studies and several clinical trials. Here, following an overview discussed autogenous/allogenic and ECM-based bone biomaterials for reconstructive surgery and applications of MSCs-mediated bone healing and tissue engineering to further offer principles and effective strategies to optimize MSCs-based bone regeneration.
Collapse
Affiliation(s)
- Fengqing Shang
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Stomatology, The 306th Hospital of PLA, Beijing, 100101, China
| | - Yang Yu
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, 250012, China
| | - Shiyu Liu
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Leiguo Ming
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yongjie Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Zhifei Zhou
- Department of Stomatology, General Hospital of Tibetan Military Command, Lhasa, 850000, China
| | - Jiayu Zhao
- Bureau of Service for Veteran Cadres of PLA in Beijing, Beijing, 100001, China
| | - Yan Jin
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| |
Collapse
|
17
|
Rabou MAA, Naga NAAE, Eid FA. Effect of Transplanted Bone Marrow on Kidney Tissue of γ-Irradiated Pregnant Rats and Their Fetuses. Pak J Biol Sci 2020; 23:92-102. [PMID: 31930887 DOI: 10.3923/pjbs.2020.92.102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
BACKGROUND AND OBJECTIVES The damaging effects of ionizing radiation lead to cell death. The present study was performed to assess the possible ameliorating effects of bone marrow transplantation (BMT) on the histopathological and histochemical changes in the kidney tissue of γ-irradiated pregnant rats and their fetuses. MATERIALS AND METHODS Pregnant rats were divided into 5 sets (6 females in each set): Group C (untreated pregnant rats), group R7 (pregnant rats exposed to 2Gy of γ-rays on the 7th day of pregnancy), group R7+BM (pregnant rats exposed to 2Gy of γ-rays on the 7th day of pregnancy then injected by freshly BMT (75×106±5 cells) intra peritoneally after 1 h of irradiation, group R14 (pregnant rats exposed to 2Gy of γ-rays on the 14th day of pregnancy), group R14+BM (pregnant rats exposed to 2Gy γ-rays on the 14th day of pregnancy and after 1 h received 1 dose of BMT). All pregnant rats were sacrificed on the 20th day of pregnancy and kidney samples of pregnant rats and their fetuses were removed for histopathological and histochemical studies. RESULTS Gamma rays caused many histological and histochemical deviations in the kidney tissue of mothers and their fetuses on day 7 or 14 of gestation, but bone marrow transplantation highly improved the damage were occurred due to γ-rays. CONCLUSION Bone marrow transplantation has the ability to decrease the injury of gamma rays.
Collapse
|
18
|
Allen ES, Conry-Cantilena C. Mobilization and collection of cells in the hematologic compartment for cellular therapies: Stem cell collection with G-CSF/plerixafor, collecting lymphocytes/monocytes. Semin Hematol 2019; 56:248-256. [PMID: 31836031 DOI: 10.1053/j.seminhematol.2019.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/01/2019] [Indexed: 12/13/2022]
Abstract
An essential and influential first step in all cellular therapies is collecting donor or patient cells. In hematopoietic progenitor cell transplantation, autologous or allogeneic hematopoietic progenitor cells (HPCs) are collected from either the bone marrow or the peripheral blood. Peripheral blood collection by apheresis requires mobilization with chemotherapy, granulocyte colony stimulating factor (G-CSF), plerixafor, or a combination. The modalities of mobilization and collection each carry a unique set of risks and benefits for both the donor and the recipient. In other types of cell therapy, most notably chimeric antigen receptor T cells, lymphocytes or monocytes are collected from the peripheral blood. The risks of collecting these cells by apheresis are similar to HPCs, but less is known about the composition, timing and qualitative cell characteristics which contribute to an optimal collection. Here, we review the mobilization and collection of HPCs and the collection of lymphocytes and monocytes. Donor safety is of primary importance when collecting material for any type of cell therapy. Every aspect of mobilization and collection can be studied and potentially optimized to improve patient outcomes.
Collapse
Affiliation(s)
- Elizabeth S Allen
- Department of Pathology, University of California San Diego, La Jolla, CA.
| | - Cathy Conry-Cantilena
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, MD
| |
Collapse
|
19
|
MacPherson A, Kimmelman J. Ethical development of stem-cell-based interventions. Nat Med 2019; 25:1037-1044. [PMID: 31270501 DOI: 10.1038/s41591-019-0511-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 06/04/2019] [Indexed: 02/07/2023]
Abstract
The process of developing new and complex stem-cell-based therapeutics is incremental and requires decades of sustained collaboration among different stakeholders. In this Perspective, we address key ethical and policy challenges confronting the clinical translation of stem-cell-based interventions (SCBIs), including premature diffusion of SCBIs to clinical practice, assessment of risk in trials, obtaining valid informed consent for research participants, balanced and complete scientific reporting and public communications, regulation, and equitable access to treatment. We propose a way forward for translating these therapies with the above challenges in mind.
Collapse
Affiliation(s)
- Amanda MacPherson
- Biomedical Ethics Unit, STREAM Research Group, McGill University, Montreal, Canada
| | - Jonathan Kimmelman
- Biomedical Ethics Unit, STREAM Research Group, McGill University, Montreal, Canada.
| |
Collapse
|
20
|
Liu Y, Han Y, Qu H, Fang J, Ye M, Yin W. Correlation of microRNA expression profile with clinical response to tumor necrosis factor inhibitor in treating rheumatoid arthritis patients: A prospective cohort study. J Clin Lab Anal 2019; 33:e22953. [PMID: 31245894 PMCID: PMC6757134 DOI: 10.1002/jcla.22953] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/24/2019] [Accepted: 05/25/2019] [Indexed: 12/14/2022] Open
Abstract
Background This study aimed to explore the correlation of circulating microRNA (miRNA) expression profile with clinical response to tumor necrosis factor (TNF) inhibitor in treating rheumatoid arthritis (RA) patients. Methods Baseline PBMC samples from eight responders and eight non‐responders after 24‐week TNF inhibitor (etanercept) treatment were subjected to miRNA microarray. Then, top 10 dysregulated miRNAs were selected and further validated by quantitative polymerase chain reaction (qPCR) in baseline PBMC samples from 92 RA patients treated with 24‐week TNF inhibitor (etanercept). Responders and non‐responders were divided referring to the decline in disease activity score in 28 joints. Results In microarray assay, total 59 upregulated and 78 downregulated miRNAs were identified in responders compared to non‐responders, which were mainly enriched in regulating immune‐ and inflammation‐related biological processes and pathways. The top 10 dysregulated miRNAs were as follows: miR‐192‐5p, miR‐146a‐5p, miR‐19b‐3p, miR‐320c, miR‐335‐5p, miR‐149‐3p, miR‐766‐3p, let‐7a‐5p, miR‐24‐3p, and miR‐1226‐5p. In qPCR validation, miR‐146a‐5p was increased, while let‐7a‐5p was decreased in responders compared with non‐responders. Multivariate logistic analysis illuminated that miR‐146a‐5p and CRP independently correlated with higher clinical response, while let‐7a‐5p and biologics history independently associated with lower clinical response. Subsequently, receiver operating characteristic curve showed that combination of these four independent factors presented with a great predictive value for clinical response with area under curve: 0.863, 95% CI 0.781‐0.945. Conclusion miRNA expression profile is closely implicated in the treatment efficacy of TNF inhibitor, and combined measurement of miR‐146a‐5p, let‐7a‐5p, CRP, and biologics history disclosed a great predictive value for clinical response to TNF inhibitor in RA patients.
Collapse
Affiliation(s)
- Yaqiong Liu
- Department of Geratology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yonghong Han
- Department of Oncology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huanru Qu
- Department of Rheumatology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingpin Fang
- Department of Rheumatology, Renji Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Mei Ye
- General Department, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wanling Yin
- Department of Geratology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Li D, Yan X, Hu Y, Liu Y, Guo R, Liao M, Shao B, Tang Q, Guo X, Chai R, Zhang Q, Tang M. Two-Photon Image Tracking of Neural Stem Cells via Iridium Complexes Encapsulated in Polymeric Nanospheres. ACS Biomater Sci Eng 2019; 5:1561-1568. [PMID: 33405629 DOI: 10.1021/acsbiomaterials.8b01231] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Iridium(III) complexes have been shown to be promising probes in two-photon imaging to real-time track the transplanted cells in stem-cell-based therapy. Here, we report on polymeric nanocapsules loaded with red phosphorescence dye of bis(2-methyldibenzo[f,h]quinoxaline) (acetylacetonate) iridium(III) (Ir(MDQ)2acac) with excellent stability created by the double emulsion method. The Ir(MDQ)2acac nanocapsules present high biocompatibility and an efficient fluorescent labeling rate when incubated with cultured mouse neural stem cells (NSCs). More importantly, the Ir(MDQ)2acac nanocapsules had both one- and two-photon imaging properties with stable phosphorescence lasting for 72 h. Furthermore, data from in vivo tracking in nude mice demonstrated that the photoluminescence from Ir(MDQ)2acac nanocapsules in NSCs could be stably monitored for up to 21 days. Our data shed light on the potential clinical application of iridium complexes encapsulated in polymeric nanospheres for two-photon imaging in real-time tracking of the transplanted stem cells.
Collapse
Affiliation(s)
- Dan Li
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.,Joint Research Institute of Southeast University and Monash University, Suzhou 215123, China
| | - Xiaoqian Yan
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.,Joint Research Institute of Southeast University and Monash University, Suzhou 215123, China
| | - Yangnan Hu
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.,Joint Research Institute of Southeast University and Monash University, Suzhou 215123, China
| | - Yun Liu
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.,Joint Research Institute of Southeast University and Monash University, Suzhou 215123, China
| | - Rongrong Guo
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.,Joint Research Institute of Southeast University and Monash University, Suzhou 215123, China
| | - Menghui Liao
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.,Joint Research Institute of Southeast University and Monash University, Suzhou 215123, China
| | - Buwei Shao
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.,Joint Research Institute of Southeast University and Monash University, Suzhou 215123, China
| | - Qilin Tang
- The First Clinical Medical School, Anhui Medical University, 81 Meishan Road, Hefei 230032, China
| | - Xing Guo
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, 211166 Jiangsu, China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.,Joint Research Institute of Southeast University and Monash University, Suzhou 215123, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing 10010, China.,ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China.,Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing 100069, China
| | - Qi Zhang
- School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Medical College of Soochow University, Suzhou, Jiangsu 215123, China
| | - Mingliang Tang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.,Joint Research Institute of Southeast University and Monash University, Suzhou 215123, China
| |
Collapse
|
22
|
General Principles of Immunotherapy in Neurological Diseases. CONTEMPORARY CLINICAL NEUROSCIENCE 2019. [DOI: 10.1007/978-3-030-19515-1_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
23
|
Analysis of Biomechanical Properties of Hematopoietic Stem and Progenitor Cells Using Real-Time Fluorescence and Deformability Cytometry. Methods Mol Biol 2019; 2017:135-148. [PMID: 31197774 DOI: 10.1007/978-1-4939-9574-5_11] [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] [Indexed: 12/30/2022]
Abstract
Stem cell mechanics, determined predominantly by the cell's cytoskeleton, plays an important role in different biological processes such as stem cell differentiation or migration. Several methods to measure mechanical properties of cells are currently available, but most of them are limited in the ability to screen large heterogeneous populations in a robust and efficient manner-a feature required for successful translational applications. With real-time fluorescence and deformability cytometry (RT-FDC), mechanical properties of cells in suspension can be screened continuously at rates of up to 1,000 cells/s-similar to conventional flow cytometers-which makes it a suitable method not only for basic research but also for a clinical setting. In parallel to mechanical characterization, RT-FDC allows to measure specific molecular markers using standard fluorescence labeling. In this chapter, we provide a detailed protocol for the characterization of hematopoietic stem and progenitor cells (HSPCs) in heterogeneous mobilized peripheral blood using RT-FDC and present a specific morpho-rheological fingerprint of HSPCs that allows to distinguish them from all other blood cell types.
Collapse
|
24
|
Raghav PK, Singh AK, Gangenahalli G. Stem cell factor and NSC87877 synergism enhances c-Kit mediated proliferation of human erythroid cells. Life Sci 2018; 214:84-97. [PMID: 30308182 DOI: 10.1016/j.lfs.2018.09.055] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/20/2018] [Accepted: 09/28/2018] [Indexed: 11/27/2022]
Abstract
The biological mechanisms underlying the effects of stem cell factor (SCF) and an inhibitor, NSC87877 (N) of the c-Kit negative regulator (SHP-1 and SHP-2) on cell proliferation are different. Therefore, we compared the cell's response to these two either alone or in combination in K562 cells. Binding of SCF (S) to c-Kit induces dimerization that activates its kinase activity. The activated c-Kit undergoes autophosphorylation at tyrosine residues that serve as a docking site for signal transduction molecules containing SH2 domains. Predominantly, the phosphotyrosine 568 (pY568) in Juxtamembrane (JM) region of c-Kit interacts with adaptor protein APS, Src family kinase, and SHP-2, while phosphotyrosine 570 (pY570) interacts with the SHP-1 and the adaptor protein Shc. The dephosphorylation of phosphotyrosine residues by SHP-1/SHP-2 leads to inhibition of c-Kit proliferative signaling. A chemical molecule, N is reported to inhibit the enzymatic activity of SHP-1/SHP-2, but its effect on c-Kit-mediated proliferation has not been studied yet. Thus, this work aims at examining the effect of the combination of S and N on cells growth as compared to individual treatment. The present study is performed with erythroleukemic K562 cells, chosen for its mRNA expression concerning the c-Kit, and SHP-1/SHP-2. Interestingly, proliferation assay showed that combination significantly increased proliferation when G1 sorted K562 cells were used. These changes were significantly higher when K562 cells were initially treated with N followed by S treatment. Collectively, these results give mechanistic insight into the proliferation enhancement of bone marrow transplantation through the synergistic effect of S and N by inhibiting SHP-1/SHP-2. The study gives solid evidence that S and N combination can be used to enhance cell proliferation/growth.
Collapse
Affiliation(s)
- Pawan Kumar Raghav
- Division of Stem Cell and Gene Therapy Research, Institute of Nuclear Medicine and Allied Sciences (INMAS), Brigadier. S. K. Mazumdar Marg, Timarpur, Delhi 110054, India
| | - Ajay Kumar Singh
- Division of Stem Cell and Gene Therapy Research, Institute of Nuclear Medicine and Allied Sciences (INMAS), Brigadier. S. K. Mazumdar Marg, Timarpur, Delhi 110054, India
| | - Gurudutta Gangenahalli
- Division of Stem Cell and Gene Therapy Research, Institute of Nuclear Medicine and Allied Sciences (INMAS), Brigadier. S. K. Mazumdar Marg, Timarpur, Delhi 110054, India.
| |
Collapse
|
25
|
Ocampo-Piraquive V, Nieto-Aristizábal I, Cañas CA, Tobón GJ. Mortality in systemic lupus erythematosus: causes, predictors and interventions. Expert Rev Clin Immunol 2018; 14:1043-1053. [DOI: 10.1080/1744666x.2018.1538789] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Vanessa Ocampo-Piraquive
- GIRAT: Grupo de Investigación en Reumatología, Autoinmunidad y Medicina traslacional, Fundación Valle del Lili and Univesidad Icesi, Cali, Colombia
| | - Ivana Nieto-Aristizábal
- GIRAT: Grupo de Investigación en Reumatología, Autoinmunidad y Medicina traslacional, Fundación Valle del Lili and Univesidad Icesi, Cali, Colombia
| | - Carlos A. Cañas
- GIRAT: Grupo de Investigación en Reumatología, Autoinmunidad y Medicina traslacional, Fundación Valle del Lili and Univesidad Icesi, Cali, Colombia
| | - Gabriel J Tobón
- GIRAT: Grupo de Investigación en Reumatología, Autoinmunidad y Medicina traslacional, Fundación Valle del Lili and Univesidad Icesi, Cali, Colombia
- Laboratory of immunology, Fundación Valle del Lili, Cali, Colombia
| |
Collapse
|
26
|
Del Papa N, Pignataro F, Zaccara E, Maglione W, Minniti A. Autologous Hematopoietic Stem Cell Transplantation for Treatment of Systemic Sclerosis. Front Immunol 2018; 9:2390. [PMID: 30386340 PMCID: PMC6198074 DOI: 10.3389/fimmu.2018.02390] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/26/2018] [Indexed: 12/29/2022] Open
Abstract
Systemic Sclerosis (SSc) is a complex autoimmune disease, characterized by high mortality and morbidity. The heterogeneity in terms of extent, severity, and rate of progression of skin and internal organ involvement gives rise to many difficulties in finding the optimal therapeutic interventions for SSc and, to date, no disease-modifying agents are available. In this scenario, it is not surprising that SSc was one of the first autoimmune diseases challenged with high-dose immunosuppressive treatment followed by autologous hematopoietic stem cell transplantation (AHSCT). In the last decades, AHSCT has emerged as a treatment option for refractory SSc through a reduction of the aberrant immune cells, followed by re-constitution of a new, self-tolerant immune system. After several case series and pilot studies, more recently three randomized controlled trials have shown a benefit in skin involvement, organ functions and quality of life measures in AHSCT compared to monthly cyclophosphamide. In addition, although AHSCT presents a certain risk of mortality, it has been shown that the overall survival is better, compared to the cyclophosphamide group. Current evidence suggests that SSc patients who are most likely to benefit from AHSCT are early, active, with rapidly progressing diffuse skin disease, and mild involvement of internal organs. As the studies have progressed, it has become evident the need for a more rigorous patient selection, the optimization of transplant and post-transplant procedures, and the intervention of multidisciplinary teams of specialists to increase the safety and efficacy of AHSCT in SSc.
Collapse
Affiliation(s)
- Nicoletta Del Papa
- Dipartimento di Fisiatria e Reumatologia, Istituto Ortopedico Gaetano Pini, Milan, Italy
| | - Francesca Pignataro
- Dipartimento di Fisiatria e Reumatologia, Istituto Ortopedico Gaetano Pini, Milan, Italy
| | - Eleonora Zaccara
- Dipartimento di Fisiatria e Reumatologia, Istituto Ortopedico Gaetano Pini, Milan, Italy
| | - Wanda Maglione
- Dipartimento di Fisiatria e Reumatologia, Istituto Ortopedico Gaetano Pini, Milan, Italy
| | - Antonina Minniti
- Dipartimento di Fisiatria e Reumatologia, Istituto Ortopedico Gaetano Pini, Milan, Italy
| |
Collapse
|
27
|
Ben Nasr M, D'Addio F, Malvandi AM, Faravelli S, Castillo-Leon E, Usuelli V, Rocchio F, Letizia T, El Essawy AB, Assi E, Mameli C, Giani E, Macedoni M, Maestroni A, Dassano A, Loretelli C, Paroni M, Cannalire G, Biasucci G, Sala M, Biffi A, Zuccotti GV, Fiorina P. Prostaglandin E2 Stimulates the Expansion of Regulatory Hematopoietic Stem and Progenitor Cells in Type 1 Diabetes. Front Immunol 2018; 9:1387. [PMID: 29971065 PMCID: PMC6018202 DOI: 10.3389/fimmu.2018.01387] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/04/2018] [Indexed: 12/29/2022] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) are multipotent stem cells that have been harnessed as a curative therapy for patients with hematological malignancies. Notably, the discovery that HSPCs are endowed with immunoregulatory properties suggests that HSPC-based therapeutic approaches may be used to treat autoimmune diseases. Indeed, infusion with HSPCs has shown promising results in the treatment of type 1 diabetes (T1D) and remains the only “experimental therapy” that has achieved a satisfactory rate of remission (nearly 60%) in T1D. Patients with newly diagnosed T1D have been successfully reverted to normoglycemia by administration of autologous HSPCs in association with a non-myeloablative immunosuppressive regimen. However, this approach is hampered by a high incidence of adverse effects linked to immunosuppression. Herein, we report that while the use of autologous HSPCs is capable of improving C-peptide production in patients with T1D, ex vivo modulation of HSPCs with prostaglandins (PGs) increases their immunoregulatory properties by upregulating expression of the immune checkpoint-signaling molecule PD-L1. Surprisingly, CXCR4 was upregulated as well, which could enhance HSPC trafficking toward the inflamed pancreatic zone. When tested in murine and human in vitro autoimmune assays, PG-modulated HSPCs were shown to abrogate the autoreactive T cell response. The use of PG-modulated HSPCs may thus provide an attractive and novel treatment of autoimmune diabetes.
Collapse
Affiliation(s)
- Moufida Ben Nasr
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States.,International Center for T1D, Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | - Francesca D'Addio
- International Center for T1D, Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | - Amir Mohammad Malvandi
- International Center for T1D, Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | - Silvia Faravelli
- International Center for T1D, Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | - Eduardo Castillo-Leon
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Vera Usuelli
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States.,International Center for T1D, Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | - Francesca Rocchio
- International Center for T1D, Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | - Teresa Letizia
- International Center for T1D, Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | | | - Emma Assi
- International Center for T1D, Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | - Chiara Mameli
- Department of Pediatrics, Buzzi Children Hospital, Milan, Italy.,Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy.,Department of Pediatrics, Children's Hospital Buzzi, Milan, Italy
| | - Elisa Giani
- Department of Pediatrics, Buzzi Children Hospital, Milan, Italy.,Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy.,Department of Pediatrics, Children's Hospital Buzzi, Milan, Italy
| | - Maddalena Macedoni
- Department of Pediatrics, Diabetes Service Studies, University of Milan, Ospedale dei Bambini Vittore Buzzi, Milan, Italy
| | - Anna Maestroni
- International Center for T1D, Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | - Alice Dassano
- International Center for T1D, Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | - Cristian Loretelli
- International Center for T1D, Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | - Moira Paroni
- Department of Bioscience, University of Milan, Milan, Italy
| | - Giuseppe Cannalire
- Department of Pediatrics and Neonatology, Ospedale Guglielmo da Saliceto, Piacenza, Italy
| | - Giacomo Biasucci
- Department of Pediatrics and Neonatology, Ospedale Guglielmo da Saliceto, Piacenza, Italy
| | - Marco Sala
- Department of Pediatrics, Tradate Hospital, Tradate, Italy
| | - Alessandra Biffi
- Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Gian Vincenzo Zuccotti
- International Center for T1D, Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy.,Department of Pediatrics, Buzzi Children Hospital, Milan, Italy.,Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy.,Department of Pediatrics, Children's Hospital Buzzi, Milan, Italy
| | - Paolo Fiorina
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States.,International Center for T1D, Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy.,Division of Endocrinology, ASST Sacco Fatebenefratelli-Sacco, Milan, Italy
| |
Collapse
|
28
|
Five year follow-up after autologous peripheral blood hematopoietic stem cell transplantation for refractory, chronic, corticosteroid-dependent systemic lupus erythematosus: effect of conditioning regimen on outcome. Bone Marrow Transplant 2018; 53:692-700. [DOI: 10.1038/s41409-018-0173-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/16/2018] [Accepted: 03/07/2018] [Indexed: 02/07/2023]
|
29
|
El-Serafi I, Remberger M, El-Serafi A, Benkessou F, Zheng W, Martell E, Ljungman P, Mattsson J, Hassan M. The effect of N-acetyl-l-cysteine (NAC) on liver toxicity and clinical outcome after hematopoietic stem cell transplantation. Sci Rep 2018; 8:8293. [PMID: 29844459 PMCID: PMC5974141 DOI: 10.1038/s41598-018-26033-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/03/2018] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED Busulphan (Bu) is a myeloablative drug used for conditioning prior to hematopoietic stem cell transplantation. Bu is predominantly metabolized through glutathione conjugation, a reaction that consumes the hepatic glutathione. N-acetyl-l-cysteine (NAC) is a glutathione precursor used in the treatment of acetaminophen hepatotoxicity. NAC does not interfere with the busulphan myeloablative effect. We investigated the effect of NAC concomitant treatment during busulphan conditioning on the liver enzymes as well as the clinical outcome. Prophylactic NAC treatment was given to 54 patients upon the start of busulphan conditioning. These patients were compared with 54 historical matched controls who did not receive NAC treatment. In patients treated with NAC, aspartate transaminase (AST), alanine transaminase (ALT) and alkaline phosphatase (ALP) were significantly (P < 0.05) decreased after conditioning compared to their start values. Within the NAC-group, liver enzymes were normalized in those patients (30%) who had significantly high start values. No significant decrease in enzyme levels was observed in the control group. Furthermore, NAC affected neither Bu kinetics nor clinical outcome (sinusoidal obstruction syndrome incidence, graft-versus-host disease and/or graft failure). IN CONCLUSION NAC is a potential prophylactic treatment for hepatotoxicity during busulphan conditioning. NAC therapy did not alter busulphan kinetics or affect clinical outcome.
Collapse
Affiliation(s)
- Ibrahim El-Serafi
- ECM, KFC, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Mats Remberger
- Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Ahmed El-Serafi
- ECM, KFC, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- College of Medicine, University of Sharjah, Sharjah, UAE
| | - Fadwa Benkessou
- ECM, KFC, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Wenyi Zheng
- ECM, KFC, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Eva Martell
- Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden
| | - Per Ljungman
- Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Mattsson
- Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Moustapha Hassan
- ECM, KFC, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.
- Experimental Cancer Medicine, Clinical Research Center, Karolinska University Hospital, Huddinge, Sweden.
| |
Collapse
|
30
|
van Megen KM, van ’t Wout EJT, Forman SJ, Roep BO. A Future for Autologous Hematopoietic Stem Cell Transplantation in Type 1 Diabetes. Front Immunol 2018; 9:690. [PMID: 29696017 PMCID: PMC5904498 DOI: 10.3389/fimmu.2018.00690] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/20/2018] [Indexed: 12/29/2022] Open
Affiliation(s)
- Kayleigh M. van Megen
- Department of Diabetes Immunology, Diabetes & Metabolism Research Institute, Beckman Research Institute at the City of Hope, Duarte, CA, United States
| | - Ernst-Jan T. van ’t Wout
- Department of Diabetes Immunology, Diabetes & Metabolism Research Institute, Beckman Research Institute at the City of Hope, Duarte, CA, United States
| | - Stephen J. Forman
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratory, City of Hope Beckman Research Institute and Medical Center, Duarte, CA, United States
| | - Bart O. Roep
- Department of Diabetes Immunology, Diabetes & Metabolism Research Institute, Beckman Research Institute at the City of Hope, Duarte, CA, United States
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
31
|
Li Z, Zhang S, Cao L, Li W, Ye YC, Shi ZX, Wang ZR, Sun LX, Wang JW, Jia LT, Wang W. Tanshinone IIA and Astragaloside IV promote the angiogenesis of mesenchymal stem cell-derived endothelial cell-like cells via upregulation of Cx37, Cx40 and Cx43. Exp Ther Med 2018; 15:1847-1854. [PMID: 29434774 PMCID: PMC5776521 DOI: 10.3892/etm.2017.5636] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 09/29/2017] [Indexed: 12/17/2022] Open
Abstract
Tanshinone IIA (Tan IIA) and Astragaloside IV (AGS-IV) were used as therapeutic treatments for coronary heart diseases (CHDs) in ancient China. However, the underlying mechanisms mediating the effects of Tan IIA and AGS-IV in angiogenesis remain unknown. In the present study, mesenchymal stem cells (MSCs) were induced to differentiate into endothelial cell (EC)-like cells in vitro and the effects of Tan IIA and/or AGS-IV on the functions of these cells, including cell proliferation and tube formation, were assessed. Compared with the single-agent groups (Tan IIA or AGS-IV only), combined-agent (Tan IIA and AGS-IV) treatment significantly enhanced the proliferation and tube formation capacity of EC-like cells. In addition, the expression of connexin 37 (Cx37), Cx40 and Cx43 in the combined-agent group was significantly increased compared with the single-agent groups. Furthermore, enhanced gap junctional intercellular communication (GJIC) was identified in the combined-agent group, as evidenced by increased dye transfer in scrape-loading dye transfer assays. In conclusion, Tan IIA and AGS-IV may promote the angiogenesis of EC-like cells by upregulating the expression of Cx37, Cx40 and Cx43 and enhancing GJIC function. The results of the present study may provide experimental evidence for the clinical application of Tan IIA and AGS-IV as a treatment for CHDs.
Collapse
Affiliation(s)
- Zhe Li
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
- Second Clinical Medical College, Shaanxi University of Traditional Chinese Medicine, Xianyang, Shaanxi 710026, P.R. China
| | - Sha Zhang
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Liang Cao
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Wei Li
- Department of Histology and Embryology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yu-Chen Ye
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Zi-Xuan Shi
- Department of Acupuncture, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi 710003, P.R. China
| | - Zong-Ren Wang
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Lian-Xu Sun
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jia-Wei Wang
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Lin-Tao Jia
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Wen Wang
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
32
|
El-Serafi I, Terelius Y, Abedi-Valugerdi M, Naughton S, Saghafian M, Moshfegh A, Mattsson J, Potácová Z, Hassan M. Flavin-containing monooxygenase 3 (FMO3) role in busulphan metabolic pathway. PLoS One 2017; 12:e0187294. [PMID: 29121650 PMCID: PMC5679629 DOI: 10.1371/journal.pone.0187294] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/17/2017] [Indexed: 12/21/2022] Open
Abstract
Busulphan (Bu) is an alkylating agent used in the conditioning regimen prior to hematopoietic stem cell transplantation (HSCT). Bu is extensively metabolized in the liver via conjugations with glutathione to form the intermediate metabolite (sulfonium ion) which subsequently is degraded to tetrahydrothiophene (THT). THT was reported to be oxidized forming THT-1-oxide that is further oxidized to sulfolane and finally 3-hydroxysulfolane. However, the underlying mechanisms for the formation of these metabolites remain poorly understood. In the present study, we performed in vitro and in vivo investigations to elucidate the involvement of flavin-containing monooxygenase-3 (FMO3) and cytochrome P450 enzymes (CYPs) in Bu metabolic pathway. Rapid clearance of THT was observed when incubated with human liver microsomes. Furthermore, among different recombinant microsomal enzymes, the highest intrinsic clearance for THT was obtained via FMO3 followed by several CYPs including 2B6, 2C8, 2C9, 2C19, 2E1 and 3A4. In Bu- or THT-treated mice, inhibition of FMO3 by phenylthiourea significantly suppressed the clearance of both Bu and THT. Moreover, the simultaneous administration of a high dose of THT (200μmol/kg) to Bu-treated mice reduced the clearance of Bu. Consistently, in patients undergoing HSCT, repeated administration of Bu resulted in a significant up-regulation of FMO3 and glutathione-S-transfrase -1 (GSTA1) genes. Finally, in a Bu-treated patient, additional treatment with voriconazole (an antimycotic drug known as an FMO3-substrate) significantly altered the Bu clearance. In conclusion, we demonstrate for the first time that FMO3 along with CYPs contribute a major part in busulphan metabolic pathway and certainly can affect its kinetics. The present results have high clinical impact. Furthermore, these findings might be important for reducing the treatment-related toxicity of Bu, through avoiding interaction with other concomitant used drugs during conditioning and hence improving the clinical outcomes of HSCT.
Collapse
Affiliation(s)
- Ibrahim El-Serafi
- Experimental Cancer Medicine (ECM), Clinical Research Centre (KFC), Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Ylva Terelius
- Experimental Cancer Medicine (ECM), Clinical Research Centre (KFC), Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Manuchehr Abedi-Valugerdi
- Experimental Cancer Medicine (ECM), Clinical Research Centre (KFC), Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Seán Naughton
- Experimental Cancer Medicine (ECM), Clinical Research Centre (KFC), Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Maryam Saghafian
- Experimental Cancer Medicine (ECM), Clinical Research Centre (KFC), Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Ali Moshfegh
- Cancer Center of Karolinska (CCK), Department of Oncology-Pathology, Karolinska Institutet, Solna, Stockholm, Sweden
| | - Jonas Mattsson
- Centre for Allogeneic Stem Cell Transplantation, Karolinska University Hospital-Huddinge, Stockholm, Sweden
- Department of Oncology and Pathology, Karolinska Institutet, Solna, Stockholm, Sweden
| | - Zuzana Potácová
- Experimental Cancer Medicine (ECM), Clinical Research Centre (KFC), Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
- ECM, Clinical Research Centre (KFC), Novum, Karolinska University Hospital, Huddinge, Sweden
| | - Moustapha Hassan
- Experimental Cancer Medicine (ECM), Clinical Research Centre (KFC), Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
- ECM, Clinical Research Centre (KFC), Novum, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
33
|
Zhang Y, Li S, Wang G, Peng Y, Zhang Q, Li H, Zhang J, Wang G, Yi S, Chen X, Xiang AP, Yang Y, Chen G. WITHDRAWN: Mesenchymal stem cells for treatment of steroid-resistant acute rejection after liver transplantation. LIVER RESEARCH 2017. [DOI: 10.1016/j.livres.2017.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
34
|
Zhang Y, Li S, Wang G, Peng Y, Zhang Q, Li H, Zhang J, Wang G, Yi S, Chen X, Xiang AP, Yang Y, Chen G. Mesenchymal stem cells for treatment of steroid-resistant acute rejection after liver transplantation. LIVER RESEARCH 2017; 1:140-145. [DOI: 10.1016/j.livres.2017.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
35
|
Vaithilingam V, Evans MDM, Lewy DM, Bean PA, Bal S, Tuch BE. Co-encapsulation and co-transplantation of mesenchymal stem cells reduces pericapsular fibrosis and improves encapsulated islet survival and function when allografted. Sci Rep 2017; 7:10059. [PMID: 28855611 PMCID: PMC5577272 DOI: 10.1038/s41598-017-10359-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 08/09/2017] [Indexed: 12/22/2022] Open
Abstract
Pericapsular fibrotic overgrowth (PFO) is associated with poor survival of encapsulated islets. A strategy to combat PFO is the use of mesenchymal stem cells (MSC). MSC have anti-inflammatory properties and their potential can be enhanced by stimulation with proinflammatory cytokines. This study investigated whether co-encapsulation or co-transplantation of MSC with encapsulated islets would reduce PFO and improve graft survival. Stimulating MSC with a cytokine cocktail of IFN-γ and TNF-α enhanced their immunosuppressive potential by increasing nitric oxide production and secreting higher levels of immunomodulatory cytokines. In vitro, co-encapsulation with MSC did not affect islet viability but significantly enhanced glucose-induced insulin secretion. In vivo, normoglycemia was achieved in 100% mice receiving islets co-encapsulated with stimulated MSC as opposed to 71.4% receiving unstimulated MSC and only 9.1% receiving encapsulated islets alone. Microcapsules retrieved from both unstimulated and stimulated MSC groups had significantly less PFO with improved islet viability and function compared to encapsulated islets alone. Levels of peritoneal immunomodulatory cytokines IL-4, IL-6, IL-10 and G-CSF were significantly higher in MSC co-encapsulated groups. Similar results were obtained when encapsulated islets and MSC were co-transplanted. In summary, co-encapsulation or co-transplantation of MSC with encapsulated islets reduced PFO and improved the functional outcome of allotransplants.
Collapse
Affiliation(s)
- Vijayaganapathy Vaithilingam
- Biomedical Manufacturing Research Program, Commonwealth Scientific and Industrial Research Organization (CSIRO), Manufacturing Flagship, North Ryde, New South Wales, Australia.
| | - Margaret D M Evans
- Biomedical Manufacturing Research Program, Commonwealth Scientific and Industrial Research Organization (CSIRO), Manufacturing Flagship, North Ryde, New South Wales, Australia
| | - Denise M Lewy
- Biomedical Manufacturing Research Program, Commonwealth Scientific and Industrial Research Organization (CSIRO), Manufacturing Flagship, North Ryde, New South Wales, Australia
| | - Penelope A Bean
- Biomedical Manufacturing Research Program, Commonwealth Scientific and Industrial Research Organization (CSIRO), Manufacturing Flagship, North Ryde, New South Wales, Australia
| | - Sumeet Bal
- Biomedical Manufacturing Research Program, Commonwealth Scientific and Industrial Research Organization (CSIRO), Manufacturing Flagship, North Ryde, New South Wales, Australia
| | - Bernard E Tuch
- Biomedical Manufacturing Research Program, Commonwealth Scientific and Industrial Research Organization (CSIRO), Manufacturing Flagship, North Ryde, New South Wales, Australia
- Australian Foundation for Diabetes Research, Sydney, New South Wales, Australia, previously at CSIRO Manufacturing Flagship, North Ryde, New South Wales, Australia
| |
Collapse
|
36
|
Xiong LL, Liu F, Deng SK, Liu J, Dan QQ, Zhang P, Zou Y, Xia QJ, Wang TH. Transplantation of Hematopoietic Stem Cells Promotes Functional Improvement Associated with NT-3-MEK-1 Activation in Spinal Cord-Transected Rats. Front Cell Neurosci 2017; 11:213. [PMID: 28769769 PMCID: PMC5515877 DOI: 10.3389/fncel.2017.00213] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 07/04/2017] [Indexed: 02/05/2023] Open
Abstract
Transected spinal cord injury (SCT) is a devastating clinical disease that strongly affects a patient’s daily life and remains a great challenge for clinicians. Stem-cell therapy has been proposed as a potential therapeutic modality for SCT. To investigate the effects of hematopoietic stem cells (HSCs) on the recovery of structure and function in SCT rats and to explore the mechanisms associated with recovery, 57 adult Sprague-Dawley rats were randomly divided into sham (n = 15), SCT (n = 24), and HSC transplantation groups (n = 15). HSCs (passage 3) labeled by Hoechst 33342, were transplanted intraspinally into the rostral, scar and caudal sites of the transected lesion at 14 days post-operation. Both in vitro and in vivo, HSCs exhibited a capacity for cell proliferation and differentiation. Following HSC transplantation, the animals’ Basso, Beattie, and Bresnahan (BBB). locomotion scale scores increased significantly between weeks 4 and 24 post-SCT, which corresponded to an increased number of 5-hydroxytryptamine (5-HT) fibers and oligodendrocytes. The amount of astrogliosis indicated by immunohistochemical staining, was markedly decreased. Moreover, the decreased expression of neurotrophin- 3 (NT-3) and mitogen-activated protein kinase kinase-1 (MEK-1) after SCT was effectively restored by HSC transplantation. The data from the current study indicate that intraspinally administered HSCs in the chronic phase of SCT results in an improvement in neurological function. Further, the results indicate that intraspinally administered HSCs benefit the underlying mechanisms involved in the enhancement of 5-HT-positive fibers and oligogenesis, the suppression of excessive astrogliosis and the upregulation of NT3-regulated MEK-1 activation in the spinal cord. These crucial findings reveal not only the mechanism of cell therapy, but may also contribute to a novel therapeutic target for the treatment of spinal cord injury (SCI).
Collapse
Affiliation(s)
- Liu-Lin Xiong
- Institute of Neurological Disease, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengdu, China
| | - Fei Liu
- Institute of Neurological Disease, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengdu, China
| | - Shi-Kang Deng
- Institute of Neuroscience, Kunming Medical UniversityKunming, China
| | - Jia Liu
- Institute of Neuroscience, Kunming Medical UniversityKunming, China
| | - Qi-Qin Dan
- Institute of Neurological Disease, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengdu, China
| | - Piao Zhang
- Institute of Neuroscience, Kunming Medical UniversityKunming, China
| | - Yu Zou
- Institute of Neurological Disease, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengdu, China
| | - Qing-Jie Xia
- Institute of Neurological Disease, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengdu, China
| | - Ting-Hua Wang
- Institute of Neurological Disease, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengdu, China.,Institute of Neuroscience, Kunming Medical UniversityKunming, China
| |
Collapse
|
37
|
Espinel JDO, Uribe C, Meyer FS, Bringheti R, Kulczynski JU, Saueressig MG. Cell therapy in the treatment of bronchiolitis obliterans in a murine model. Rev Col Bras Cir 2017; 42:181-8. [PMID: 26291260 DOI: 10.1590/0100-69912015003010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 10/10/2014] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE To evaluate the importance of stem cells derived from adipose tissue in reducing graft inflammation in a murine model of allogeneic heterotopic tracheal transplant. METHODS We performed a heterotopic tracheal allografting in dorsal subcutaneous pouch and systemically injected 5x105 mesenchymal stem cells derived from adipose tissue. The animals were divided into two groups according to the time of sacrifice: T7 and T21. We also carried out histological analysis and digital morphometry. RESULTS The T7 animals treated with cell therapy had median obstructed graft area of 0 versus 0.54 of controls (p = 0.635). The treated T21 subjects had median obstructed graft area of 0.25 versus 0 in controls (p = 0.041). CONCLUSION The systemically injected cell therapy in experimental murine model of bronchiolitis obliterans did not reduce the severity of the allograft inflammation in a statistically significant way in seven days; Conversely, in 21 days, it increased the allograft inflammatory process.
Collapse
Affiliation(s)
| | | | | | - Rafael Bringheti
- Universidade Federal de Ciências da Saúde de Porto Alegre, RS, BR
| | | | | |
Collapse
|
38
|
Abstract
Autoimmune dacryoadenitis, such as Sjögren syndrome, comprises multifactorial and complex diseases. Inflammation of the lacrimal gland plays a key role in the pathogenesis of diseases. Unfortunately, current treatment strategies, including artificial tears, anti-inflammatory drugs, punctual occlusion, and immunosuppressive drugs, are only palliative, and long-term administration of these strategies is associated with adverse effects that limit their utility. Hence, an effective and safe treatment for autoimmune dacryoadenitis is urgently needed. Mesenchymal stem cells (MSCs) have emerged as a promising tool for treating autoimmune dacryoadenitis, owing to their immunosuppressive properties, tissue repair functions, and powerful differentiation capabilities. A large number of studies have focused on the effect of MSCs on autoimmune diseases, such as autoimmune uveitis, inflammatory bowel disease, and collagen-induced arthritis, but few studies have, to date, unequivocally established the efficacy of MSCs for treating autoimmune dacryoadenitis. In this review, we discuss recent advances in MSC treatment for autoimmune dacryoadenitis.
Collapse
Affiliation(s)
- Xiaoxiao Lu
- Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute & Tianjin Medical University School of Optometry and Ophthalmology, No.251 Fukang Road, Nankai District, Tianjin, 300384, People's Republic of China
| | - Xilian Wang
- Tianjin Beichen Hospital, No. 7, Beiyi Road, Beichen District, Tianjin, 300400, China
| | - Hong Nian
- Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute & Tianjin Medical University School of Optometry and Ophthalmology, No.251 Fukang Road, Nankai District, Tianjin, 300384, People's Republic of China
| | - Dan Yang
- Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute & Tianjin Medical University School of Optometry and Ophthalmology, No.251 Fukang Road, Nankai District, Tianjin, 300384, People's Republic of China
| | - Ruihua Wei
- Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute & Tianjin Medical University School of Optometry and Ophthalmology, No.251 Fukang Road, Nankai District, Tianjin, 300384, People's Republic of China.
| |
Collapse
|
39
|
Zhou J, Cui H, Lu H, Xu Z, Feng W, Chen L, Jin X, Yang X, Qi Z. Muscle-derived stem cells in peripheral nerve regeneration: reality or illusion? Regen Med 2017. [PMID: 28621200 DOI: 10.2217/rme-2016-0165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Owing to the complicated and time-consuming regenerative process, the repair of injured peripheral nerves depends largely on ongoing stem-cell therapy. Decades ago, researchers successfully isolated and identified muscle-derived stem cells (MDSCs) and discovered their potential for multidifferentiation. MDSCs play an important role in trauma repair associated with neuromuscular and vascular injury by simultaneously promoting tissue regrowth via direct differentiation and systematic secretion under physiological conditions. However, the isolation, culture, induction and application of MDSCs require further methodological analysis before clinical application. In this review, we comprehensively discuss the challenges associated with neural regeneration and reviewed the progress of stem cell based regenerative medicine, in an effort to realize the potential of MDSCs in nerve regeneration.
Collapse
Affiliation(s)
- Jing Zhou
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| | - Haiyan Cui
- Department of Plastic & Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China
| | - Haibin Lu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| | - Zhuqiu Xu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| | - Weifeng Feng
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| | - Lulu Chen
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| | - Xiaolei Jin
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| | - Xiaonan Yang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| | - Zuoliang Qi
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| |
Collapse
|
40
|
Abstract
Cell therapy is considered a promising potential treatment for multiple sclerosis, perhaps particularly for the progressive form of the disease for which there are currently no useful treatments. Over the past two decades or more, much progress has been made in understanding the biology of MS and in the experimental development of cell therapy for this disease. Three quite distinct forms of cell therapy are currently being pursued. The first seeks to use stem cells to replace damaged myelin-forming oligodendrocytes within the CNS; the second aims, in effect, to replace the individual's misfunctioning immune system, making use of haematopoietic stem cells; and the third seeks to utilise endogenous stem cell populations by mobilisation with or without in vitro expansion, exploiting their various reparative and neuroprotective properties. In this article we review progress in these three separate areas, summarising the experimental background and clinical progress thus far made.
Collapse
|
41
|
Stem-cell transplantation for Crohn's disease: same authors, different conclusions? Lancet Gastroenterol Hepatol 2017; 2:386-387. [DOI: 10.1016/s2468-1253(17)30076-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 03/06/2017] [Indexed: 11/15/2022]
|
42
|
Blair NF, Frith TJR, Barbaric I. Regenerative Medicine: Advances from Developmental to Degenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1007:225-239. [PMID: 28840560 DOI: 10.1007/978-3-319-60733-7_12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chronic tissue and organ failure caused by an injury, disease, ageing or congenital defects represents some of the most complex therapeutic challenges and poses a significant financial healthcare burden. Regenerative medicine strategies aim to fulfil the unmet clinical need by restoring the normal tissue function either through stimulating the endogenous tissue repair or by using transplantation strategies to replace the missing or defective cells. Stem cells represent an essential pillar of regenerative medicine efforts as they provide a source of progenitors or differentiated cells for use in cell replacement therapies. Whilst significant leaps have been made in controlling the stem cell fates and differentiating them to cell types of interest, transitioning bespoke cellular products from an academic environment to off-the-shelf clinical treatments brings about a whole new set of challenges which encompass manufacturing, regulatory and funding issues. Notwithstanding the need to resolve such issues before cell replacement therapies can benefit global healthcare, mounting progress in the field has highlighted regenerative medicine as a realistic prospect for treating some of the previously incurable conditions.
Collapse
Affiliation(s)
- Nicholas F Blair
- Wellcome Trust - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Thomas J R Frith
- Centre for Stem Cell Biology, Department of Biomedical Science, The University of Sheffield, Sheffield, UK
| | - Ivana Barbaric
- Centre for Stem Cell Biology, Department of Biomedical Science, The University of Sheffield, Sheffield, UK.
| |
Collapse
|
43
|
Liao L, Shi B, Chang H, Su X, Zhang L, Bi C, Shuai Y, Du X, Deng Z, Jin Y. Heparin improves BMSC cell therapy: Anticoagulant treatment by heparin improves the safety and therapeutic effect of bone marrow-derived mesenchymal stem cell cytotherapy. Theranostics 2017; 7:106-116. [PMID: 28042320 PMCID: PMC5196889 DOI: 10.7150/thno.16911] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 09/14/2016] [Indexed: 12/15/2022] Open
Abstract
Systemic infusion of bone marrow-derived mesenchymal stem cells (BMSCs) has become a promising strategy for disease treatment and tissue regeneration. Strategies to enhance the efficiency of BMSC cell therapy are crucial to promote its clinical application. Here, we aimed to improve BMSC cell therapy by inhibiting the BMSC-induced coagulation reaction. Intravenous injection of gradient BMSCs into mice showed that BMSCs were not fully compatible with blood. Large doses of BMSCs induced a series of symptoms of respiratory failure and heart failure. Histological and homeostasis analysis confirmed that large doses of BMSCs induced disseminated intravascular thrombosis, exhaustion of platelets and coagulation factors, and prolonged prothrombin time (PT) and activated partial thromboplastin time (APTT). Similar to mouse BMSCs, goat and human BMSCs also induced coagulation reactions in vitro and in vivo. The coagulation was induced mostly by tissue factor, the overexpression of which enhanced the procoagulant activity of BMSCs during in vitro culture. Notably, clinical doses of BMSCs in cell therapy also induced mild and reversible coagulation, which increased BMSC lung embolism and clearance. Anticoagulation treatment by heparin (400 U/kg) prevented BMSC-induced coagulation and the acute adverse effects of large-dose BMSCs infusion efficiently. Importantly, heparin treatment led to decreased BMSC lung embolism and enhanced migration and maintenance of BMSCs to target organs in cell therapy. Based on an experimental colitis model, we confirmed that heparin treatment enhanced the effect of BMSC therapy efficiently to reduce mortality, prevent weight loss, suppress inflammation reaction and alleviate tissue injury. In conclusion, BMSCs possess procoagulant activity that could induce disseminated coagulation and thrombosis in recipients. Anticoagulation treatment by heparin is a practical strategy to improve both the safety and therapeutic effect of BMSC therapy.
Collapse
|
44
|
Yu Y, Liao L, Shao B, Su X, Shuai Y, Wang H, Shang F, Zhou Z, Yang D, Jin Y. Knockdown of MicroRNA Let-7a Improves the Functionality of Bone Marrow-Derived Mesenchymal Stem Cells in Immunotherapy. Mol Ther 2016; 25:480-493. [PMID: 28153095 DOI: 10.1016/j.ymthe.2016.11.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 11/03/2016] [Accepted: 11/27/2016] [Indexed: 02/06/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) have been recently used in clinical treatment of inflammatory diseases. Practical strategies improving the immunosuppressive property of MSCs are urgently needed for MSC immunotherapy. In this study, we aimed to develop a microRNA-based strategy to improve MSC immunotherapy. Bioinformatic analysis revealed that let-7a targeted the 3' UTR of mRNA of Fas and FasL, both of which are essential for MSCs to induce T cell apoptosis. Knockdown of let-7a by specific inhibitor doubled Fas and Fas ligand (FasL) protein levels in MSCs. Because Fas attracts T cell migration and FasL induces T cell apoptosis, knockdown of let-7a significantly promoted MSC-induced T cell migration and apoptosis in vitro and in vivo. Importantly, MSCs knocked down of let-7a were more efficient to reduce the mortality, prevent the weight loss, suppress the inflammation reaction, and alleviate the tissue lesion of experimental colitis and graft-versus-host disease (GVHD) mouse models. In conclusion, knockdown of let-7a significantly improved the therapeutic effect of MSC cytotherapy on inflammatory bowel diseases and GVHD. With high safety and convenience, knockdown of let-7a is a potential strategy to improve MSC therapy for inflammatory diseases in clinic.
Collapse
Affiliation(s)
- Yang Yu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China; Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Li Liao
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China; Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi 710032, China; State Key Laboratory of Military Stomatology, Department of Oral Histology and Pathology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Bingyi Shao
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China; Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Xiaoxia Su
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yi Shuai
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China; Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi 710032, China; State Key Laboratory of Military Stomatology, Department of Oral Histology and Pathology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Han Wang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China; Department of Stomatology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510000, China
| | - Fengqing Shang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China; Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi 710032, China; State Key Laboratory of Military Stomatology, Department of Oral Histology and Pathology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zhifei Zhou
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China; Department of Stomatology, General Hospital of Tibetan Military Region, Lasa, Tibet 850000, China
| | - Deqin Yang
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China.
| | - Yan Jin
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China; Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
45
|
Symonds G, Bartlett JS, Kiem HP, Tsie M, Breton L. Cell-Delivered Entry Inhibitors for HIV-1: CCR5 Downregulation and Blocking Virus/Membrane Fusion in Defending the Host Cell Population. AIDS Patient Care STDS 2016; 30:545-550. [PMID: 27905841 DOI: 10.1089/apc.2016.0245] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
HIV-1 infection requires the presence of the CD4 receptor on the target cell surface and a coreceptor, predominantly CC-chemokine receptor 5 (CCR5). It has been shown that individuals who are homozygous for a defective CCR5 gene are protected from HIV-1 infection. A novel self-inactivating lentiviral vector LVsh5/C46 (Cal-1) has been engineered to block HIV-1 infection with two viral entry inhibitors, conferring resistance to HIV-1 infection from both CCR5 and CXCR4 tropic strains. Cal-1 encodes a short hairpin RNA (sh5) to downregulate CCR5 and C46, an HIV-1 fusion inhibitor. Gene therapy by Cal-1 is aimed at transducing CD4+ T cells and CD34+ hematopoietic stem/progenitor cells in an autologous transplant setting. Pre-clinical safety and efficacy studies in vitro and in vivo (humanized mouse model and nonhuman primates) have shown that Cal-1 is safe with no indication of any toxicity risk and acts to decrease viral load and increase CD4 counts. Two clinical trials are underway using Cal-1: a phase I/II study to assess safety and feasibility in an adult HIV-1-positive population not on antiretroviral therapy (ART); and a second Fred Hutchinson Investigator Initiated phase I study to assess safety and feasibility in adults with HIV-1-associated non-Hodgkin or Hodgkin lymphoma.
Collapse
|
46
|
Abstract
Pediatrics is a dynamic discipline and there is awareness and hope for actualizing outstanding achievements in the field of child health in 21st century and beyond. Improved lifestyle and quality of children's health is likely to reduce the burden of adult diseases and enhance longevity because seeds of most adult diseases are sown in childhood. Identification and decoding of human genome is expected to revolutionize the practice of pediatrics. The day is not far off when a patient will walk into doctor's chamber with an electronic or digital health history on a CD or palmtop and a decoded genomic constitution. There will be reduced burden of genetic diseases because of selective abortions of "defective" fetuses and replacement of "bad" genes with "good" ones by genetic engineering. Availability of totipotent stem cells and developments in transplant technology are likely to revolutionize the management of a variety of hematologic cancers and life-threatening genetic disorders. The possibility of producing flawless designer babies by advances in assisted reproductive technologies (ARTs) is likely to be mired by several ethical and legal issues.The availability of newer vaccines by recombinant technology for emerging infective and for non-infective lifestyle diseases is likely to improve survival and quality of life. There is going to be a greater focus on the "patient" having the disease rather than "disease" per se by practicing holistic pediatrics by effective utilization of alternative or complementary strategies for health care. Due to advances in technology, pediatrics may get further dehumanized. A true healer cannot simply rely on technology; there must be a spiritual bond between the patient and the physician by exploiting the concept of psycho-neuro-immunology and body-mind interactions. In the years to come, physicians are likely to play "god" but medicine can't achieve immortality because anything born must die in accordance with nature's recycling blueprint. The medical science is likely to improve longevity but our goal should be to improve the quality of life.
Collapse
Affiliation(s)
- Meharban Singh
- Child Care Center, 625, Sector 37, Arun Vihar, Noida, UP, India.
| |
Collapse
|
47
|
Ginoux E, Kottler D, Anglaret B, Balme B, Bulabois CE, Skowron F. Remission of a long-lasting sarcoidosis after allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia. JAAD Case Rep 2016; 2:408-410. [PMID: 27766304 PMCID: PMC5065641 DOI: 10.1016/j.jdcr.2016.08.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Emmanuelle Ginoux
- Department of Dermatology, Centre Hospitalier de Valence, Valence, France
| | - Diane Kottler
- Department of Dermatology, Centre Hospitalier de Valence, Valence, France
| | - Bruno Anglaret
- Department of Hematology, Centre Hospitalier de Valence, Valence, France
| | - Brigitte Balme
- Department of Dermatopathology, CHU Lyon sud, Pierre Bénite, France
| | | | - François Skowron
- Department of Dermatology, Centre Hospitalier de Valence, Valence, France
- Correspondence to: François Skowron, MD, Service de Dermatologie, Centre Hospitalier de Valence, 179 Bd Maréchal JUIN, 26000 Valence, France.Service de DermatologieCentre Hospitalier de Valence179 Bd Maréchal JUINValence26000France
| |
Collapse
|
48
|
The Current State of Cell Therapies for Cerebrovascular Diseases. Stem Cells Int 2016; 2016:5215824. [PMID: 27672396 PMCID: PMC5031840 DOI: 10.1155/2016/5215824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 08/21/2016] [Indexed: 11/17/2022] Open
|
49
|
Wingstrand VL, Grønhøj Larsen C, Jensen DH, Bork K, Sebbesen L, Balle J, Fischer-Nielsen A, von Buchwald C. Mesenchymal Stem Cell Therapy for the Treatment of Vocal Fold Scarring: A Systematic Review of Preclinical Studies. PLoS One 2016; 11:e0162349. [PMID: 27631373 PMCID: PMC5025194 DOI: 10.1371/journal.pone.0162349] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 08/22/2016] [Indexed: 01/02/2023] Open
Abstract
Objectives Therapy with mesenchymal stem cells exhibits potential for the development of novel interventions for many diseases and injuries. The use of mesenchymal stem cells in regenerative therapy for vocal fold scarring exhibited promising results to reduce stiffness and enhance the biomechanical properties of injured vocal folds. This study evaluated the biomechanical effects of mesenchymal stem cell therapy for the treatment of vocal fold scarring. Data Sources PubMed, Embase, the Cochrane Library and Google Scholar were searched. Methods Controlled studies that assessed the biomechanical effects of mesenchymal stem cell therapy for the treatment of vocal fold scarring were included. Primary outcomes were viscoelastic properties and mucosal wave amplitude. Results Seven preclinical animal studies (n = 152 single vocal folds) were eligible for inclusion. Evaluation of viscoelastic parameters revealed a decreased dynamic viscosity (η’) and elastic modulus (G’), i.e., decreased resistance and stiffness, in scarred vocal folds treated with mesenchymal stem cells compared to non-treated scarred vocal folds. Mucosal wave amplitude was increased in scarred vocal folds treated with mesenchymal stem cells vs. non-treated scarred vocal folds. Conclusion The results from these studies suggest an increased regenerative effect of therapy with mesenchymal stem cells for scarred vocal folds and are encouraging for further clinical studies.
Collapse
Affiliation(s)
- Vibe Lindeblad Wingstrand
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Christian Grønhøj Larsen
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| | - David H. Jensen
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Bork
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lars Sebbesen
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Balle
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anne Fischer-Nielsen
- Cell Therapy Facility, Blood Bank, Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Christian von Buchwald
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
50
|
Li Z, Rubinstein SM, Thota R, Savani M, Brissot E, Shaw BE, Majhail NS, Mohty M, Savani BN. Immune-Mediated Complications after Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2016; 22:1368-1375. [PMID: 27095688 DOI: 10.1016/j.bbmt.2016.04.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 04/06/2016] [Indexed: 11/27/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) has an integral role in the treatment of malignant and nonmalignant diseases. Long-term complications after HSCT have been well established and include graft-versus-host disease (GVHD), conditioning regimen-related toxicities, disease relapse, and infections. Immune-mediated phenomena are increasingly described after HSCT with clinically significant sequelae. Diagnosis is challenging because of features that overlap with other commonly reported post-transplantation complications. Patients who experience immune-mediated disease after HSCT tend to have poor outcomes. Early recognition of immune-mediated complications is imperative to reduce preventable morbidity and mortality. This review looks at the currently available literature on pathogenesis, incidence, risk factors, treatment, and outcomes of immune-mediated disease (other than GVHD) after HSCT.
Collapse
Affiliation(s)
- Zhuoyan Li
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Samuel M Rubinstein
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ramya Thota
- Hematology and Stem Cell Transplantation Section, Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center and Veterans Affairs Medical Center, Nashville, Tennessee
| | - Malvi Savani
- University of Tennessee Heath Science Center, College of Medicine, Memphis, Tennessee
| | - Eolia Brissot
- Department of Haematology, Saint Antoine Hospital, Paris, France; INSERM UMR 938, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Bronwen E Shaw
- Center for International Blood and Marrow Transplant Research and Froedtert and the Medical College of Wisconsin, Wisconsin
| | - Navneet S Majhail
- Blood and Marrow Transplant Program, Cleveland Clinic, Cleveland, Ohio
| | - Mohamad Mohty
- Department of Haematology, Saint Antoine Hospital, Paris, France; INSERM UMR 938, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Bipin N Savani
- Hematology and Stem Cell Transplantation Section, Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center and Veterans Affairs Medical Center, Nashville, Tennessee.
| |
Collapse
|