101
|
Kaltenborn A, Nolte A, Schwager Y, Littbarski SA, Emmanouilidis N, Arelin V, Klempnauer J, Schrem H. Identification of patients at risk for renal impairment after living donor kidney transplantation. Langenbecks Arch Surg 2016; 401:1219-1229. [PMID: 27502290 DOI: 10.1007/s00423-016-1492-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 07/29/2016] [Indexed: 12/31/2022]
Abstract
PURPOSE Outcome after living donor kidney transplantation is highly relevant, since recipient and donor were exposed to notable harm. Reliable identification of risk factors is necessary. METHODS Three hundred sixty-six living donor kidney transplants were included in this observational retrospective study. Relevant risk factors for renal impairment 1 year after transplantation and delayed graft function were identified with univariable and multivariable binary logistic regression and ordinal regression analysis. RESULTS Eighty-four patients (26.6 %) suffered from renal impairment KDIGO stage ≥4 1 year post-transplant; median estimated glomerular filtration rate was 35.3 ml/min. In multivariable ordinal regression, male recipient sex (p < 0.001), recipient body mass index (p = 0.006), donor age (p = 0.002) and high percentages of panel reactive antibodies (p = 0.021) were revealed as independent risk factors for higher KDIGO stages. After adjustment for post-transplant data, recipient male sex (p < 0.001), donor age (p = 0.026) and decreased early renal function at the first post-transplant outpatient visit (p < 0.001) were identified as independent risk factors. Delayed graft function was independently associated with long stay on the waiting list (p = 0.011), high donor body mass index (p = 0.043), prolonged warm ischemic time (p = 0.016) and the presence of preformed donor-specific antibodies (p = 0.043). CONCLUSIONS Broadening the donor pool with non-blood related donors seems to be legitimate, although with respect to careful medical selection, since donor age in combination with male recipient sex were shown to be risk factors for decreased graft function. Warm ischemic time and waiting time need to be kept as short as possible to avoid delayed graft function. Transplantation across HLA and ABO borders did not affect outcome significantly.
Collapse
Affiliation(s)
- Alexander Kaltenborn
- Core Facility Quality Management and Health Technology Assessment in Transplantation, Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany. .,Department of Trauma and Orthopaedic Surgery, Federal Armed Forces Hospital Westerstede, Westerstede, Germany.
| | - Almut Nolte
- Core Facility Quality Management and Health Technology Assessment in Transplantation, Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany.,Ministry of Defence, Bonn, Germany
| | - Ysabell Schwager
- Core Facility Quality Management and Health Technology Assessment in Transplantation, Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Simon A Littbarski
- Core Facility Quality Management and Health Technology Assessment in Transplantation, Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Nikos Emmanouilidis
- Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Viktor Arelin
- Core Facility Quality Management and Health Technology Assessment in Transplantation, Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany.,Department of Nephrology, Hannover Medical School, Hannover, Germany
| | - Jürgen Klempnauer
- Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Harald Schrem
- Core Facility Quality Management and Health Technology Assessment in Transplantation, Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany.,Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| |
Collapse
|
102
|
Davies LC, Alm JJ, Heldring N, Moll G, Gavin C, Batsis I, Qian H, Sigvardsson M, Nilsson B, Kyllonen LE, Salmela KT, Carlsson PO, Korsgren O, Le Blanc K. Type 1 Diabetes Mellitus Donor Mesenchymal Stromal Cells Exhibit Comparable Potency to Healthy Controls In Vitro. Stem Cells Transl Med 2016; 5:1485-1495. [PMID: 27412884 DOI: 10.5966/sctm.2015-0272] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 04/01/2016] [Indexed: 12/13/2022] Open
Abstract
: Bone marrow mesenchymal stromal cells (BM-MSCs) have been characterized and used in many clinical studies based on their immunomodulatory and regenerative properties. We have recently reported the benefit of autologous MSC systemic therapy in the treatment of type 1 diabetes mellitus (T1D). Compared with allogeneic cells, use of autologous products reduces the risk of eliciting undesired complications in the recipient, including rejection, immunization, and transmission of viruses and prions; however, comparable potency of autologous cells is required for this treatment approach to remain feasible. To date, no analysis has been reported that phenotypically and functionally characterizes MSCs derived from newly diagnosed and late-stage T1D donors in vitro with respect to their suitability for systemic immunotherapy. In this study, we used gene array in combination with functional in vitro assays to address these questions. MSCs from T1D donors and healthy controls were expanded from BM aspirates. BM mononuclear cell counts and growth kinetics were comparable between the groups, with equivalent colony-forming unit-fibroblast capacity. Gene microarrays demonstrated differential gene expression between healthy and late-stage T1D donors in relation to cytokine secretion, immunomodulatory activity, and wound healing potential. Despite transcriptional differences, T1D MSCs did not demonstrate a significant difference from healthy controls in immunosuppressive activity, migratory capacity, or hemocompatibility. We conclude that despite differential gene expression, expanded MSCs from T1D donors are phenotypically and functionally similar to healthy control MSCs with regard to their immunomodulatory and migratory potential, indicating their suitability for use in autologous systemic therapy. SIGNIFICANCE The potential for mesenchymal stromal cells (MSCs) as a cell-based therapy in the treatment of immunologic disorders has been well established. Recent studies reported the clinical potential for autologous MSCs as a systemic therapy in the treatment of type I diabetes mellitus (T1D). The current study compared the genotypic and phenotypic profiles of bone marrow-derived MSCs from T1D and healthy donors as autologous (compared with allogeneic) therapy provides distinct advantages, such as reduced risk of immune reaction and transmission of infectious agents. The findings of the current study demonstrate that despite moderate differences in T1D MSCs at the gene level, these cells can be expanded in culture to an extent corresponding to that of MSCs derived from healthy donors. No functional difference in terms of immunosuppressive activity, blood compatibility, or migratory capacity was evident between the groups. The study findings also show that autologous MSC therapy holds promise as a T1D treatment and should be evaluated further in clinical trials.
Collapse
Affiliation(s)
- Lindsay C Davies
- Center for Hematology and Regenerative Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Divisions of Clinical Immunology and Transfusion Medicine, Department of Laboratory Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Jessica J Alm
- Center for Hematology and Regenerative Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Divisions of Clinical Immunology and Transfusion Medicine, Department of Laboratory Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Nina Heldring
- Center for Hematology and Regenerative Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Divisions of Clinical Immunology and Transfusion Medicine, Department of Laboratory Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Guido Moll
- Center for Hematology and Regenerative Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Divisions of Clinical Immunology and Transfusion Medicine, Department of Laboratory Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Caroline Gavin
- Center for Hematology and Regenerative Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Divisions of Clinical Immunology and Transfusion Medicine, Department of Laboratory Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Ioannis Batsis
- Center for Hematology and Regenerative Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Hong Qian
- Center for Hematology and Regenerative Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Mikael Sigvardsson
- Institution for Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Bo Nilsson
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Sweden
| | - Lauri E Kyllonen
- Division of Transplantation, Helsinki University Hospital, Helsinki, Finland
| | - Kaija T Salmela
- Division of Transplantation, Helsinki University Hospital, Helsinki, Finland
| | - Per-Ola Carlsson
- Department of Medical Cell Biology, Uppsala University, Sweden
- Department of Medical Sciences, Uppsala University, Sweden
| | - Olle Korsgren
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Sweden
| | - Katarina Le Blanc
- Center for Hematology and Regenerative Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Divisions of Clinical Immunology and Transfusion Medicine, Department of Laboratory Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
103
|
Massie AB, Leanza J, Fahmy LM, Chow EKH, Desai NM, Luo X, King EA, Bowring MG, Segev DL. A Risk Index for Living Donor Kidney Transplantation. Am J Transplant 2016; 16:2077-84. [PMID: 26752290 PMCID: PMC6114098 DOI: 10.1111/ajt.13709] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 11/20/2015] [Accepted: 12/13/2015] [Indexed: 01/25/2023]
Abstract
Choosing between multiple living kidney donors, or evaluating offers in kidney paired donation, can be challenging because no metric currently exists for living donor quality. Furthermore, some deceased donor (DD) kidneys can result in better outcomes than some living donor kidneys, yet there is no way to compare them on the same scale. To better inform clinical decision-making, we created a living kidney donor profile index (LKDPI) on the same scale as the DD KDPI, using Cox regression and adjusting for recipient characteristics. Donor age over 50 (hazard ratio [HR] per 10 years = 1.15 1.241.33 ), elevated BMI (HR per 10 units = 1.01 1.091.16 ), African-American race (HR = 1.15 1.251.37 ), cigarette use (HR = 1.09 1.161.23 ), as well as ABO incompatibility (HR = 1.03 1.271.58 ), HLA B (HR = 1.03 1.081.14 ) mismatches, and DR (HR = 1.04 1.091.15 ) mismatches were associated with greater risk of graft loss after living donor transplantation (all p < 0.05). Median (interquartile range) LKDPI score was 13 (1-27); 24.2% of donors had LKDPI < 0 (less risk than any DD kidney), and 4.4% of donors had LKDPI > 50 (more risk than the median DD kidney). The LKDPI is a useful tool for comparing living donor kidneys to each other and to deceased donor kidneys.
Collapse
Affiliation(s)
- A B Massie
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Epidemiology, Johns Hopkins School of Public Health, Baltimore, MD
| | - J Leanza
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - L M Fahmy
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - E K H Chow
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - N M Desai
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - X Luo
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - E A King
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - M G Bowring
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - D L Segev
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Epidemiology, Johns Hopkins School of Public Health, Baltimore, MD
- Scientific Registry for Transplant Recipients, Minneapolis, MN
| |
Collapse
|
104
|
Kowalski TJ, Leong NL, Dar A, Wu L, Kabir N, Khan AZ, Eliasberg CD, Pedron A, Karayan A, Lee S, Di Pauli von Treuheim T, Jiacheng J, Wu BM, Evseenko D, McAllister DR, Petrigliano FA. Hypoxic culture conditions induce increased metabolic rate and collagen gene expression in ACL-derived cells. J Orthop Res 2016; 34:985-94. [PMID: 26621359 DOI: 10.1002/jor.23116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 11/25/2015] [Indexed: 02/04/2023]
Abstract
There has been substantial effort directed toward the application of bone marrow and adipose-derived mesenchymal stromal cells (MSCs) in the regeneration of musculoskeletal tissue. Recently, resident tissue-specific stem cells have been described in a variety of mesenchymal structures including ligament, tendon, muscle, cartilage, and bone. In the current study, we systematically characterize three novel anterior cruciate ligament (ACL)-derived cell populations with the potential for ligament regeneration: ligament-forming fibroblasts (LFF: CD146(neg) , CD34(neg) CD44(pos) , CD31(neg) , CD45(neg) ), ligament perivascular cells (LPC: CD146(pos) CD34(neg) CD44(pos) , CD31(neg) , CD45(neg) ) and ligament interstitial cells (LIC: CD34(pos) CD146(neg) , CD44(pos) , CD31(neg) , CD45(neg) )-and describe their proliferative and differentiation potential, collagen gene expression and metabolism in both normoxic and hypoxic environments, and their trophic potential in vitro. All three groups of cells (LIC, LPC, and LFF) isolated from adult human ACL exhibited progenitor cell characteristics with regard to proliferation and differentiation potential in vitro. Culture in low oxygen tension enhanced the collagen I and III gene expression in LICs (by 2.8- and 3.3-fold, respectively) and LFFs (by 3- and 3.5-fold, respectively) and increased oxygen consumption rate and extracellular acidification rate in LICs (by 4- and 3.5-fold, respectively), LFFs (by 5.5- and 3-fold, respectively), LPCs (by 10- and 4.5-fold, respectively) as compared to normal oxygen concentration. In summary, this study demonstrates for the first time the presence of three novel progenitor cell populations in the adult ACL that demonstrate robust proliferative and matrix synthetic capacity; these cells may play a role in local ligament regeneration, and consequently represent a potential cell source for ligament engineering applications. © 2015 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:985-994, 2016.
Collapse
Affiliation(s)
- Tomasz J Kowalski
- Department of Orthopaedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, 90095, California
| | - Natalie L Leong
- Department of Orthopaedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, 90095, California
| | - Ayelet Dar
- Department of Orthopaedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, 90095, California
| | - Ling Wu
- Department of Orthopaedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, 90095, California
| | - Nima Kabir
- Department of Orthopaedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, 90095, California
| | - Adam Z Khan
- Department of Orthopaedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, 90095, California
| | - Claire D Eliasberg
- Department of Orthopaedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, 90095, California
| | - Andrew Pedron
- Department of Orthopaedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, 90095, California
| | - Anthony Karayan
- Department of Orthopaedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, 90095, California
| | - Siyoung Lee
- Department of Orthopaedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, 90095, California
| | - Theodor Di Pauli von Treuheim
- Department of Orthopaedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, 90095, California
| | - Jin Jiacheng
- Department of Orthopaedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, 90095, California
| | - Ben M Wu
- Department of Bioengineering, University of California, Los Angeles, 90095, California
| | - Denis Evseenko
- Department of Orthopaedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, 90095, California
| | - David R McAllister
- Department of Orthopaedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, 90095, California
| | - Frank A Petrigliano
- Department of Orthopaedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, 90095, California
| |
Collapse
|
105
|
Li B, Cairns JA, Robb ML, Johnson RJ, Watson CJE, Forsythe JL, Oniscu GC, Ravanan R, Dudley C, Roderick P, Metcalfe W, Tomson CR, Bradley JA. Predicting patient survival after deceased donor kidney transplantation using flexible parametric modelling. BMC Nephrol 2016; 17:51. [PMID: 27225846 PMCID: PMC4881185 DOI: 10.1186/s12882-016-0264-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 05/16/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The influence of donor and recipient factors on outcomes following kidney transplantation is commonly analysed using Cox regression models, but this approach is not useful for predicting long-term survival beyond observed data. We demonstrate the application of a flexible parametric approach to fit a model that can be extrapolated for the purpose of predicting mean patient survival. The primary motivation for this analysis is to develop a predictive model to estimate post-transplant survival based on individual patient characteristics to inform the design of alternative approaches to allocating deceased donor kidneys to those on the transplant waiting list in the United Kingdom. METHODS We analysed data from over 12,000 recipients of deceased donor kidney or combined kidney and pancreas transplants between 2003 and 2012. We fitted a flexible parametric model incorporating restricted cubic splines to characterise the baseline hazard function and explored a range of covariates including recipient, donor and transplant-related factors. RESULTS Multivariable analysis showed the risk of death increased with recipient and donor age, diabetic nephropathy as the recipient's primary renal diagnosis and donor hypertension. The risk of death was lower in female recipients, patients with polycystic kidney disease and recipients of pre-emptive transplants. The final model was used to extrapolate survival curves in order to calculate mean survival times for patients with specific characteristics. CONCLUSION The use of flexible parametric modelling techniques allowed us to address some of the limitations of both the Cox regression approach and of standard parametric models when the goal is to predict long-term survival.
Collapse
Affiliation(s)
- Bernadette Li
- Department of Health Services Research and Policy, London School of Hygiene and Tropical Medicine, 15-17 Tavistock Place, London, WC1H 9SH, UK.
| | - John A Cairns
- Department of Health Services Research and Policy, London School of Hygiene and Tropical Medicine, 15-17 Tavistock Place, London, WC1H 9SH, UK
| | | | | | - Christopher J E Watson
- Department of Surgery, University of Cambridge and the NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - John L Forsythe
- Transplant Unit, Royal Infirmary of Edinburgh, Edinburgh, UK
| | | | - Rommel Ravanan
- Richard Bright Renal Unit, Southmead Hospital, Bristol, UK
| | | | - Paul Roderick
- Primary Care and Population Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Charles R Tomson
- Department of Renal Medicine, Freeman Hospital, Newcastle upon Tyne, UK
| | - J Andrew Bradley
- Department of Surgery, University of Cambridge and the NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| |
Collapse
|
106
|
Utilizing Autologous Multipotent Mesenchymal Stromal Cells and β-Tricalcium Phosphate Scaffold in Human Bone Defects: A Prospective, Controlled Feasibility Trial. BIOMED RESEARCH INTERNATIONAL 2016; 2016:2076061. [PMID: 27144159 PMCID: PMC4838782 DOI: 10.1155/2016/2076061] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/09/2016] [Indexed: 12/28/2022]
Abstract
The purpose of this prospective controlled study was to compare healing quality following the implantation of ultraporous β-tricalcium phosphate, containing either expanded autologous mesenchymal stromal cells (trial group, 9 patients) or β-tricalcium phosphate alone (control group, 9 patients), into femoral defects during revision total hip arthroplasty. Both groups were assessed using the Harris Hip Score, radiography, and DEXA scanning at 6 weeks and 3, 6, and 12 months postoperatively. A significant difference in the bone defect healing was observed between both groups of patients (P < 0.05). In the trial group, trabecular remodeling was found in all nine patients and in the control group, in 1 patient only. Whereas, over the 12-month follow-up period, no significant difference was observed between both groups of patients in terms of the resorption of β-tricalcium phosphate, the significant differences were documented in the presence of radiolucency and bone trabeculation through the defect (P < 0.05). Using autologous mesenchymal stromal cells combined with a β-tricalcium phosphate scaffold is a feasible, safe, and effective approach for management of bone defects with compromised microenvironment. The clinical trial was registered at the EU Clinical Trials Register before patient recruitment has begun (EudraCT number 2012-005599-33).
Collapse
|
107
|
Mehrfarjam Z, Esmaeili F, Shabani L, Ebrahimie E. Induction of pancreatic β cell gene expression in mesenchymal stem cells. Cell Biol Int 2016; 40:486-500. [DOI: 10.1002/cbin.10567] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 11/23/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Zahra Mehrfarjam
- Razi Herbal Medicines Research Center; Lorestan University of Medical Sciences; P.O. Box 681499468 Khorramabad Iran
| | - Fariba Esmaeili
- Faculty of Basic Sciences; Department of Biology; University of Isfahan; P.O. Box 8174673441 Isfahan Iran
- Research Institute of Biotechnology; Shahrekord University; P.O. Box 115 Shahrekord Iran
| | - Leila Shabani
- Research Institute of Biotechnology; Shahrekord University; P.O. Box 115 Shahrekord Iran
| | - Esmaeil Ebrahimie
- Institute of Biotechnology; Shiraz University; Shiraz Iran
- Division of Information Technology, Engineering & Environment; School of Information Technology and Mathematical Sciences; University of South Australia; Adelaide Australia
- Department of Genetics and Evolution; The University of Adelaide; Adelaide Australia
- Faculty of Science and Engineering; School of Biological Sciences; Flinders University; Adelaide Australia
| |
Collapse
|
108
|
Prologo JD, Hawkins M, Gilliland C, Chinnadurai R, Harkey P, Chadid T, Lee Z, Brewster L. Interventional stem cell therapy. Clin Radiol 2016; 71:307-11. [PMID: 26874660 DOI: 10.1016/j.crad.2016.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/26/2015] [Accepted: 01/04/2016] [Indexed: 12/13/2022]
Abstract
The ability to deliver cells in appropriate doses to their targeted site of action is a well-known obstacle to optimising stem cell therapy. Systemic administration of cells results in pulmonary "trapping," which significantly decreases the number of available circulating cells to impact underlying disorders. Directed delivery of stem cells in interventional radiology may provide an additional option for bypassing the lungs, as well as introduce novel potential avenues for decreasing doses required to effect cellular therapy, efficiently obtain local paracrine effects, and/or to simplify targeting strategies.
Collapse
Affiliation(s)
- J D Prologo
- Division of Interventional Radiology and Image Guided Medicine, Emory University School of Medicine, 1364 Clifton Rd NE, Suite AG05, Atlanta, GA 30322, USA.
| | - M Hawkins
- Division of Interventional Radiology and Image Guided Medicine, Emory University School of Medicine, 1364 Clifton Rd NE, Suite AG05, Atlanta, GA 30322, USA
| | - C Gilliland
- Division of Interventional Radiology and Image Guided Medicine, Emory University School of Medicine, 1364 Clifton Rd NE, Suite AG05, Atlanta, GA 30322, USA
| | - R Chinnadurai
- Department of Hematology and Oncology, Emory University School of Medicine, Winship Cancer Institute, 1365B Clifton Rd NE, Suite B506, Atlanta, GA 30322, USA
| | - P Harkey
- Division of Musculoskeletal Radiology, Emory University School of Medicine, 1364 Clifton Rd NE, Suite AG05, Atlanta, GA 30322, USA
| | - T Chadid
- Department of Surgery, Emory University School of Medicine, 1364 Clifton Rd NE, Suite H100, Atlanta, GA 30322, USA
| | - Z Lee
- Department of Radiology, Case Western Reserve University College of Medicine, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| | - Luke Brewster
- Department of Surgery, Emory University School of Medicine, 1364 Clifton Rd NE, Suite H100, Atlanta, GA 30322, USA; Department of Surgical and Research Services, Atlanta Veterans Medical Center, 1670 Clairmont Road, Decatur, GA 30033, USA
| |
Collapse
|
109
|
Staels W, De Groef S, Heremans Y, Coppens V, Van Gassen N, Leuckx G, Van de Casteele M, Van Riet I, Luttun A, Heimberg H, De Leu N. Accessory cells for β-cell transplantation. Diabetes Obes Metab 2016; 18:115-24. [PMID: 26289770 DOI: 10.1111/dom.12556] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/22/2015] [Accepted: 08/13/2015] [Indexed: 12/16/2022]
Abstract
Despite recent advances, insulin therapy remains a treatment, not a cure, for diabetes mellitus with persistent risk of glycaemic alterations and life-threatening complications. Restoration of the endogenous β-cell mass through regeneration or transplantation offers an attractive alternative. Unfortunately, signals that drive β-cell regeneration remain enigmatic and β-cell replacement therapy still faces major hurdles that prevent its widespread application. Co-transplantation of accessory non-islet cells with islet cells has been shown to improve the outcome of experimental islet transplantation. This review will highlight current travails in β-cell therapy and focuses on the potential benefits of accessory cells for islet transplantation in diabetes.
Collapse
MESH Headings
- Animals
- Cell Proliferation
- Cell Separation/trends
- Cells, Cultured
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/surgery
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/surgery
- Endothelial Progenitor Cells/cytology
- Endothelial Progenitor Cells/immunology
- Endothelial Progenitor Cells/pathology
- Endothelial Progenitor Cells/transplantation
- Graft Rejection/immunology
- Graft Rejection/metabolism
- Graft Rejection/prevention & control
- Graft Survival
- Humans
- Immune Tolerance
- Insulin-Secreting Cells/cytology
- Insulin-Secreting Cells/immunology
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/transplantation
- Islets of Langerhans Transplantation/adverse effects
- Islets of Langerhans Transplantation/immunology
- Mesenchymal Stem Cell Transplantation/adverse effects
- Mesenchymal Stem Cell Transplantation/trends
- Neural Crest/cytology
- Neural Crest/immunology
- Neural Crest/pathology
- Neural Crest/transplantation
- Stem Cell Transplantation/adverse effects
- Stem Cell Transplantation/trends
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- T-Lymphocytes, Regulatory/transplantation
- Transplantation, Autologous/adverse effects
- Transplantation, Autologous/trends
- Transplantation, Heterotopic/adverse effects
- Transplantation, Heterotopic/trends
- Transplantation, Homologous/adverse effects
- Transplantation, Homologous/trends
Collapse
Affiliation(s)
- W Staels
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- Division of Pediatric Endocrinology, Department of Pediatrics, Ghent University Hospital, Ghent, Belgium
- Department of Pediatrics and Genetics, Ghent University, Ghent, Belgium
| | - S De Groef
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Y Heremans
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - V Coppens
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - N Van Gassen
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - G Leuckx
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - M Van de Casteele
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - I Van Riet
- Department Hematology Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - A Luttun
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - H Heimberg
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - N De Leu
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Endocrinology, UZ Brussel, Brussels, Belgium
- Department of Endocrinology, ASZ Aalst, Aalst, Belgium
| |
Collapse
|
110
|
Wan Y, Garner J, Wu N, Phillip L, Han Y, McDaniel K, Annable T, Zhou T, Francis H, Glaser S, Huang Q, Alpini G, Meng F. Role of stem cells during diabetic liver injury. J Cell Mol Med 2016; 20:195-203. [PMID: 26645107 PMCID: PMC4727564 DOI: 10.1111/jcmm.12723] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 09/24/2015] [Indexed: 12/18/2022] Open
Abstract
Diabetes mellitus is one of the most severe endocrine metabolic disorders in the world that has serious medical consequences with substantial impacts on the quality of life. Type 2 diabetes is one of the main causes of diabetic liver diseases with the most common being non-alcoholic fatty liver disease. Several factors that may explain the mechanisms related to pathological and functional changes of diabetic liver injury include: insulin resistance, oxidative stress and endoplasmic reticulum stress. The realization that these factors are important in hepatocyte damage and lack of donor livers has led to studies concentrating on the role of stem cells (SCs) in the prevention and treatment of liver injury. Possible avenues that the application of SCs may improve liver injury include but are not limited to: the ability to differentiate into pancreatic β-cells (insulin producing cells), the contribution for hepatocyte regeneration, regulation of lipogenesis, glucogenesis and anti-inflammatory actions. Once further studies are performed to explore the underlying protective mechanisms of SCs and the advantages and disadvantages of its application, there will be a greater understand of the mechanism and therapeutic potential. In this review, we summarize the findings regarding the role of SCs in diabetic liver diseases.
Collapse
Affiliation(s)
- Ying Wan
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
- Academic Operations, Baylor Scott & White Healthcare, Temple, TX, USA
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research, Southern Medical University, Guangzhou, China
| | - Jessica Garner
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
- Academic Operations, Baylor Scott & White Healthcare, Temple, TX, USA
| | - Nan Wu
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
| | - Levine Phillip
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
- Academic Operations, Baylor Scott & White Healthcare, Temple, TX, USA
| | - Yuyan Han
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
| | - Kelly McDaniel
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
- Academic Operations, Baylor Scott & White Healthcare, Temple, TX, USA
| | - Tami Annable
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Academic Operations, Baylor Scott & White Healthcare, Temple, TX, USA
| | - Tianhao Zhou
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
- Academic Operations, Baylor Scott & White Healthcare, Temple, TX, USA
| | - Heather Francis
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
- Academic Operations, Baylor Scott & White Healthcare, Temple, TX, USA
| | - Shannon Glaser
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
| | - Qiaobing Huang
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research, Southern Medical University, Guangzhou, China
| | - Gianfranco Alpini
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
| | - Fanyin Meng
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
- Academic Operations, Baylor Scott & White Healthcare, Temple, TX, USA
| |
Collapse
|
111
|
Tao H, Han Z, Han ZC, Li Z. Proangiogenic Features of Mesenchymal Stem Cells and Their Therapeutic Applications. Stem Cells Int 2016; 2016:1314709. [PMID: 26880933 PMCID: PMC4736816 DOI: 10.1155/2016/1314709] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 11/04/2015] [Accepted: 11/29/2015] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have shown their therapeutic potency for treatment of cardiovascular diseases owing to their low immunogenicity, ease of isolation and expansion, and multipotency. As multipotent progenitors, MSCs have revealed their ability to differentiate into various cell types and could promote endogenous angiogenesis via microenvironmental modulation. Studies on cardiovascular diseases have demonstrated that transplanted MSCs could engraft at the injured sites and differentiate into cardiomyocytes and endothelial cells as well. Accordingly, several clinical trials using MSCs have been performed and revealed that MSCs may improve relevant clinical parameters in patients with vascular diseases. To fully comprehend the characteristics of MSCs, understanding their intrinsic property and associated modulations in tuning their behaviors as well as functions is indispensable for future clinical translation of MSC therapy. This review will focus on recent progresses on endothelial differentiation and potential clinical application of MSCs, with emphasis on therapeutic angiogenesis for treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Hongyan Tao
- Department of Pathophysiology, Nankai University School of Medicine, Tianjin 300071, China
- The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University College of Life Science, Tianjin 300071, China
| | - Zhibo Han
- State Key Lab of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Zhong Chao Han
- State Key Lab of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Zongjin Li
- Department of Pathophysiology, Nankai University School of Medicine, Tianjin 300071, China
- The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University College of Life Science, Tianjin 300071, China
| |
Collapse
|
112
|
Rackham CL, Vargas AE, Hawkes RG, Amisten S, Persaud SJ, Austin ALF, King AJF, Jones PM. Annexin A1 Is a Key Modulator of Mesenchymal Stromal Cell-Mediated Improvements in Islet Function. Diabetes 2016; 65:129-39. [PMID: 26470781 DOI: 10.2337/db15-0990] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 10/07/2015] [Indexed: 11/13/2022]
Abstract
We have previously demonstrated that coculture of islets with mesenchymal stromal cells (MSCs) enhanced islet insulin secretory capacity in vitro, correlating with improved graft function in vivo. To identify factors that contribute to MSC-mediated improvements in islet function, we have used an unbiased quantitative RT-PCR screening approach to identify MSC-derived peptide ligands of G-protein-coupled receptors that are expressed by islets cells. We demonstrated high expression of annexin A1 (ANXA1) mRNA by MSCs and confirmed expression at the protein level in lysates and MSC-conditioned media by Western blot analysis and ELISA. Preculturing islets with exogenous ANXA1 enhanced glucose-stimulated insulin secretion (GSIS), thereby mimicking the beneficial influence of MSC preculture in vitro. Small interfering RNA-mediated knockdown of ANXA1 in MSCs reduced their capacity to potentiate GSIS. MSCs derived from ANXA1(-/-) mice had no functional capacity to enhance GSIS, in contrast to wild-type controls. Preculturing islets with ANXA1 had modest effects on their capacity to regulate blood glucose in streptozotocin-induced diabetic mice, indicating that additional MSC-derived factors are required to fully mimic the beneficial effects of MSC preculture in vivo. These findings demonstrate the feasibility of harnessing the MSC secretome as a defined, noncellular strategy to improve the efficiency of clinical islet transplantation protocols.
Collapse
Affiliation(s)
- Chloe L Rackham
- Diabetes Research Group, Division of Diabetes & Nutritional Sciences, School of Medicine, King's College London, London, U.K
| | - Andreia E Vargas
- Diabetes Research Group, Division of Diabetes & Nutritional Sciences, School of Medicine, King's College London, London, U.K
| | - Ross G Hawkes
- Diabetes Research Group, Division of Diabetes & Nutritional Sciences, School of Medicine, King's College London, London, U.K
| | - Stefan Amisten
- Diabetes Research Group, Division of Diabetes & Nutritional Sciences, School of Medicine, King's College London, London, U.K
| | - Shanta J Persaud
- Diabetes Research Group, Division of Diabetes & Nutritional Sciences, School of Medicine, King's College London, London, U.K
| | - Amazon L F Austin
- Diabetes Research Group, Division of Diabetes & Nutritional Sciences, School of Medicine, King's College London, London, U.K
| | - Aileen J F King
- Diabetes Research Group, Division of Diabetes & Nutritional Sciences, School of Medicine, King's College London, London, U.K
| | - Peter M Jones
- Diabetes Research Group, Division of Diabetes & Nutritional Sciences, School of Medicine, King's College London, London, U.K.
| |
Collapse
|
113
|
Hypoxic preconditioning potentiates the trophic effects of mesenchymal stem cells on co-cultured human primary hepatocytes. Stem Cell Res Ther 2015; 6:237. [PMID: 26626568 PMCID: PMC4667488 DOI: 10.1186/s13287-015-0218-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 10/14/2015] [Accepted: 10/28/2015] [Indexed: 12/16/2022] Open
Abstract
Introduction Mesenchymal stem/stromal cells (MSCs) improve the metabolic function of co-cultured hepatocytes. The present study aimed to further enhance the trophic effects of co-culture with hepatocytes using hypoxic preconditioning (HPc) of the MSCs and also to investigate the underlying molecular mechanisms involved. Methods Human adipose tissue-derived MSCs were subjected to hypoxia (2 % O2; HPc) or normoxia (20 % O2) for 24 h and then co-cultured with isolated human hepatocytes. Assays of metabolic function and apoptosis were performed to investigate the hepatotrophic and anti-apoptotic effects of co-culture. Indirect co-cultures and co-culture with MSC-conditioned medium investigated the role of paracrine factors in the hepatotrophic effects of co-culture. Reactive oxygen species (ROS) activity was antagonised with N-acetylcysteine to investigate whether HPc potentiated the effects of MSCs by intracellular ROS-dependent mechanisms. Tumour necrosis factor (TNF)-α, transforming growth factor (TGF)-β1, and extracellular collagen production was determined and CASP9 and BAX/BCL-2 signalling pathways analysed to investigate the role of soluble factors, extracellular matrix deposition, and apoptosis-associated gene signalling in the effects of co-culture. Results HPc potentiated the hepatotrophic and anti-apoptotic effects of co-culture by ROS-dependent mechanisms. There was increased MSC TGF-β1 production, and enhanced MSC deposition of extracellular collagen, with reduced synthesis of TNF-α, as well as a downregulation of the expression of pro-apoptotic CASP9, BAX, BID and BLK genes and upregulated expression of anti-apoptotic BCL-2 in hepatocytes. Conclusions HPc potentiated the trophic and anti-apoptotic effects of MSCs on hepatocytes via mechanisms including intracellular ROS, autocrine TGF-β, extracellular collagen and caspase and BAX/BCL-2 signalling pathways. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0218-7) contains supplementary material, which is available to authorized users.
Collapse
|
114
|
Cao S, Wei X, Li H, Miao J, Zhao G, Wu D, Liu B, Zhang Y, Gu H, Wang L, Fan Y, An D, Yuan Z. Comparative Study on the Differentiation of Mesenchymal Stem Cells Between Fetal and Postnatal Rat Spinal Cord Niche. Cell Transplant 2015; 25:1115-30. [PMID: 26651539 DOI: 10.3727/096368915x689910] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In a previous study, we established a prenatal surgical approach and transplanted mesenchymal stem cells (MSCs) into the fetal rat spinal column to treat neural tube defects (NTDs). We found that the transplanted MSCs survived and differentiated into neural lineage cells. Various cytokines and extracellular signaling systems in the spinal cord niche play an important role in cell differentiation. In this study, we observed the differentiation of transplanted MSCs in different spinal cord niches and further observed the expression of neurotrophic factors and growth factors in the spinal cord at different developmental stages to explore the mechanism of MSC differentiation in different spinal cord niches. The results showed that transplanted MSCs expressed markers of neural precursor cells (nestin), neurogliocytes (GFAP), and neurons (β-tubulin). The percentages of GFP(+)/nestin(+) double-positive cells in transplanted MSCs in E16, P1, and P21 rats were 18.31%, 12.18%, and 5.06%, respectively. The percentages of GFP(+)/GFAP(+) double-positive cells in E16, P1, and P21 rats were 32.01%, 15.35%, and 12.56%, respectively. The percentages of GFP(+)/β-tubulin(+) double-positive cells in E16, P1, and P21 were 11.76%, 7.62%, and 4.88%, respectively. The differentiation rates of MSCs in embryonic spinal cords were significantly higher than in postnatal spinal cords (p < 0.05). We found that the transplanted MSCs expressed synapsin-1 at different developmental stages. After MSC transplantation, we observed that neurotrophic factor-3 (NT-3), fibroblast growth factor-2 (FGF-2), FGF-8, transforming growth factor-α (TGF-α), vascular endothelial growth factor (VEGF), and platelet-derived growth factor (PDGF) significantly increased in the MSC transplantation group compared with the blank injection group. Furthermore, FGF-2 and VEGF expression were positively correlated with the number of surviving MSCs. In addition, we found that the expression of brain-derived neurotrophic factor (BDNF), NT-3, FGF-8, TGF-β, epidermal growth factor (EGF), and insulin-like growth factor (IGF) decreased with age, and the expression of FGF-2, FGF-10, FGF-20, TGF-α, and PDGF increased with age. Our data suggest that the embryonic spinal cord niche is more conducive to MSC differentiation after transplantation.
Collapse
Affiliation(s)
- Songying Cao
- Department of Pediatric Surgery, Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
115
|
El-Demerdash RF, Hammad LN, Kamal MM, El Mesallamy HO. A comparison of Wharton's jelly and cord blood as a source of mesenchymal stem cells for diabetes cell therapy. Regen Med 2015; 10:841-855. [PMID: 26541176 DOI: 10.2217/rme.15.49] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM In this study, we investigated the differences between mesenchymal stem cells (MSCs), isolated from umbilical cord blood (UCB-MSCs) and Wharton's jelly (WJ-MSCs) as sources of diabetes mellitus cell therapy. METHODS After isolation, both cell types were induced to differentiate into insulin producing cells, then the differentiated cells were assessed genetically and functionally. UCB-MSCs and WJ-MSCs were transplanted in the tail veins of streptozotocin-induced diabetic rats. Blood glucose levels were monitored post-transplantation. RESULTS & CONCLUSION Wharton's jelly was more homogeneous, can better differentiate into insulin producing cells in vitro and better control hyperglycemia in diabetic rats in vivo, as compared with UCB. These results indicate that WJ-MSCs represent a potential source of cells in the field of diabetes mellitus cell therapy.
Collapse
Affiliation(s)
- Rasha F El-Demerdash
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Misr International University, Cairo, Egypt, 44971
| | - Lamiaa N Hammad
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Misr International University, Cairo, Egypt, 44971
| | - Mohamed M Kamal
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt, 11566
| | - Hala O El Mesallamy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt, 11566
| |
Collapse
|
116
|
Li J, Peng X, Zeng X, Liu B, Hao Q, Yu X, Zhu L, Hu Q. Estrogen Secreted by Mesenchymal Stem Cells Necessarily Determines Their Feasibility of Therapeutical Application. Sci Rep 2015; 5:15286. [PMID: 26478095 PMCID: PMC5389885 DOI: 10.1038/srep15286] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 09/22/2015] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells are therapeutically applicable and involved in the development of some types of diseases including estrogen (E2)-related ones. Little is known about E2 secretion by mesenchymal stem cells and its potential influence on their therapeutical applications. Our in vitro experiments showed that BMSCs cultured from C57BL/6J mice secreted E2 in a time-dependent manner. In vivo study identified a significantly increased E2 level in serum after a single administration of BMSCs, and a sustained elevation of E2 level upon a repetitive administration. Morris water maze test in the ovariectomised (OVX) mouse model revealed BMSCs transplantation ameliorated OVX-induced memory deficits by secreted E2. On the contrary, in endometriosis model, BMSCs transplantation aggravated endometriotic lesions because of E2 secretion. Mechanistically, the aromatase cytochrome P450 appeared to be critical for the biosynthesis and exerted effects of estrogen secretion by BMSCs. Our findings suggested that BMSCs transplantation is on the one hand an attractive option for the therapeutic treatment of diseases associated with E2 deficits in part through E2 secretion, on the other hand a detrimental factor for the E2-exasperated diseases largely via E2 production. It is important and necessary to monitor serum E2 level before and after the initiation of BMSCs therapy.
Collapse
Affiliation(s)
- Jiansha Li
- Key Laboratory of Pulmonary Diseases of Ministry of Health and Department of Pathophysiology, School of Basic Medicine
- Department of Pathology, Tongji Hospital; Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430030, People’s Republic of China
| | - Xiaochun Peng
- Key Laboratory of Pulmonary Diseases of Ministry of Health and Department of Pathophysiology, School of Basic Medicine
| | - Xianqin Zeng
- Key Laboratory of Pulmonary Diseases of Ministry of Health and Department of Pathophysiology, School of Basic Medicine
| | - Bingxun Liu
- Key Laboratory of Pulmonary Diseases of Ministry of Health and Department of Pathophysiology, School of Basic Medicine
| | - Qiang Hao
- Key Laboratory of Pulmonary Diseases of Ministry of Health and Department of Pathophysiology, School of Basic Medicine
| | - Xiangyuan Yu
- Key Laboratory of Pulmonary Diseases of Ministry of Health and Department of Pathophysiology, School of Basic Medicine
| | - Liping Zhu
- Key Laboratory of Pulmonary Diseases of Ministry of Health and Department of Pathophysiology, School of Basic Medicine
| | - Qinghua Hu
- Key Laboratory of Pulmonary Diseases of Ministry of Health and Department of Pathophysiology, School of Basic Medicine
| |
Collapse
|
117
|
Ben Nasr M, Vergani A, Avruch J, Liu L, Kefaloyianni E, D'Addio F, Tezza S, Corradi D, Bassi R, Valderrama-Vasquez A, Usuelli V, Kim J, Azzi J, El Essawy B, Markmann J, Abdi R, Fiorina P. Co-transplantation of autologous MSCs delays islet allograft rejection and generates a local immunoprivileged site. Acta Diabetol 2015; 52:917-27. [PMID: 25808641 PMCID: PMC4968999 DOI: 10.1007/s00592-015-0735-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 03/05/2015] [Indexed: 12/29/2022]
Abstract
AIMS Mesenchymal stem cells (MSCs) are multipotent cells with immunomodulatory properties. We tested the ability of MSCs to delay islet allograft rejection. METHODS Mesenchymal stem cells were generated in vitro from C57BL/6 and BALB/c mice bone marrow, and their immunomodulatory properties were tested in vitro. We then tested the effect of a local or systemic administration of heterologous and autologous MSCs on graft survival in a fully allogeneic model of islet transplantation (BALB/c islets into C57BL/6 mice). RESULTS In vitro, autologous, but not heterologous, MSCs abrogated immune cell proliferation in response to alloantigens and skewed the immune response toward a Th2 profile. A single dose of autologous MSCs co-transplanted under the kidney capsule with allogeneic islets delayed islet rejection, reduced graft infiltration, and induced long-term graft function in 30 % of recipients. Based on ex vivo analysis of recipient splenocytes, the use of autologous MSCs did not appear to have any systemic effect on the immune response toward graft alloantigens. The systemic injection of autologous MSCs or the local injection of heterologous MSCs failed to delay islet graft rejection. CONCLUSION Autologous, but not heterologous, MSCs showed multiple immunoregulatory properties in vitro and delayed allograft rejection in vivo when co-transplanted with islets; however, they failed to prevent rejection when injected systemically. Autologous MSCs thus appear to produce a local immunoprivileged site, which promotes graft survival.
Collapse
Affiliation(s)
- Moufida Ben Nasr
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Enders Building 5th Floor Room EN511, 300 Longwood Ave, Boston, MA, USA
- Transplant Medicine, Ospedale San Raffaele, Milan, Italy
| | - Andrea Vergani
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Enders Building 5th Floor Room EN511, 300 Longwood Ave, Boston, MA, USA
- Transplant Medicine, Ospedale San Raffaele, Milan, Italy
| | - James Avruch
- Transplantation Unit, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Liye Liu
- Transplantation Unit, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Eirini Kefaloyianni
- Renal Division, Brigham and Women's Hospital, Harvard Institute of Medicine, HIM510, Harvard Medical School, Boston, MA, 02115, USA
| | - Francesca D'Addio
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Enders Building 5th Floor Room EN511, 300 Longwood Ave, Boston, MA, USA
- Transplant Medicine, Ospedale San Raffaele, Milan, Italy
| | - Sara Tezza
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Enders Building 5th Floor Room EN511, 300 Longwood Ave, Boston, MA, USA
| | - Domenico Corradi
- Pathology and Laboratory Medicine, University of Parma, Parma, Italy
| | - Roberto Bassi
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Enders Building 5th Floor Room EN511, 300 Longwood Ave, Boston, MA, USA
| | | | - Vera Usuelli
- Transplant Medicine, Ospedale San Raffaele, Milan, Italy
| | - James Kim
- Transplantation Unit, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Jamil Azzi
- Nephrology Division, Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - James Markmann
- Transplantation Unit, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Reza Abdi
- Nephrology Division, Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Paolo Fiorina
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Enders Building 5th Floor Room EN511, 300 Longwood Ave, Boston, MA, USA.
- Transplant Medicine, Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
118
|
Fransson M, Brännström J, Duprez I, Essand M, Le Blanc K, Korsgren O, Magnusson PU. Mesenchymal stromal cells support endothelial cell interactions in an intramuscular islet transplantation model. Regen Med Res 2015; 3:1. [PMID: 26430512 PMCID: PMC4589952 DOI: 10.1186/s40340-015-0010-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 09/18/2015] [Indexed: 12/13/2022] Open
Abstract
Background Mesenchymal stromal cells (MSC) have been under investigation for a number of therapies and have lately been in focus as immunosuppressive actors in the field of transplantation. Herein we have extended our previously published in vitro model of MSC-islets in an experimental setting of islet transplantation to the abdominal muscle. Human islets coated with luciferase-GFP transduced human MSC were transplanted to the abdomen muscle tissue of NOD-scid ILR2γnull mice and cellular interactions were investigated by confocal microscopy. Results The MSC reduced fibrotic encapsulation and facilitated endothelial cell interactions. In particular, we show a decreased fraction of αSMA expressing fibrotic tissue surrounding the graft in presence of MSC-islets compared to islets solely distributed into the muscle tissue. Also, in the presence of MSC, human islet endothelial cells migrated from the center of the graft out into the surrounding tissue forming chimeric blood vessels with recipient endothelial cells. Further, in the graft periphery, MSC were seen interacting with infiltrating macrophages. Conclusions Here, in our experimental in vivo model of composite human islets and luciferase-GFP-transduced human MSC, we enable the visualization of close interactions between the MSC and the surrounding tissue. In this model of transplantation the MSC contribute to reduced fibrosis and increased islet endothelial cell migration. Furthermore, the MSC interact with the recipient vasculature and infiltrating macrophages. Electronic supplementary material The online version of this article (doi:10.1186/s40340-015-0010-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Moa Fransson
- Department of Immunology, Genetics and Pathology, Division of Clinical Immunology, Uppsala, Sweden
| | - Johan Brännström
- Department of Immunology, Genetics and Pathology, Division of Clinical Immunology, Uppsala, Sweden
| | - Ida Duprez
- Department of Immunology, Genetics and Pathology, Division of Clinical Immunology, Uppsala, Sweden
| | - Magnus Essand
- Department of Immunology, Genetics and Pathology, Division of Clinical Immunology, Uppsala, Sweden
| | - Katarina Le Blanc
- Department of Clinical Immunology and Transfusion Medicine, Karolinska Institutet and Hematology Center at Karolinska University Hospital, Huddinge, Sweden
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Division of Clinical Immunology, Uppsala, Sweden
| | - Peetra U Magnusson
- Department of Immunology, Genetics and Pathology, Division of Clinical Immunology, Uppsala, Sweden ; Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Dag Hammarskjölds Väg 20, SE-751 85 Uppsala, Sweden
| |
Collapse
|
119
|
Päth G, Seufert J. Stammzelltherapie des Diabetes mellitus Typ 1. DIABETOLOGE 2015. [DOI: 10.1007/s11428-015-0024-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
120
|
Bone marrow-derived mesenchymal stem cells ameliorate chronic high glucose-induced β-cell injury through modulation of autophagy. Cell Death Dis 2015; 6:e1885. [PMID: 26379190 PMCID: PMC4650435 DOI: 10.1038/cddis.2015.230] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/07/2015] [Accepted: 06/18/2015] [Indexed: 12/13/2022]
Abstract
Chronic hyperglycemia causes a progressive decrease of β-cell function and mass in type 2 diabetic patients. Growing evidence suggests that augment of autophagy may be an effective approach to protect β cells against various extra-/intracellular stimuli. In this study, we thus investigated whether bone marrow-derived mesenchymal stem cells (BM-MSCs) could ameliorate chronic high glucose (HG)-induced β-cell injury through modulation of autophagy. Prolonged exposure to HG decreased cell viability, increased cell apoptosis and impaired basal insulin secretion and glucose-stimulated insulin secretion of INS-1 cells, but BM-MSC treatment significantly alleviated these glucotoxic alternations. In addition, western blotting displayed upregulated expression of Beclin1 and LC3-II in INS-1 cells co-cultured with BM-MSCs. Results from immunofluorescence staining and transmission electronic microscope analysis also revealed that BM-MSCs promoted autophagosomes and autolysosomes formation in HG-treated INS-1 cells. However, it should be noted that inhibition of autophagy significantly diminished the protective effects of BM-MSCs on HG-treated INS-1 cells, suggesting that the improvement of β-cell function and survival induced by BM-MSCs was mediated through autophagy. Furthermore, our results showed that BM-MSCs improved mitochondrial function and reduced reactive oxygen species production in HG-treated INS-1 cells, largely owing to autophagic clearance of impaired mitochondria. In vivo study was performed in rats with type 2 diabetes (T2D). BM-MSC infusion not only ameliorated hyperglycemia, but also promoted restoration of pancreatic β cells in T2D rats. Meanwhile, BM-MSC infusion upregulated LAMP2 expression and enhanced formation of autophagosomes and autolysosomes, combined with reduced β-cell apoptosis and increased number of insulin granules. These findings together indicated that BM-MSCs could protect β cells against chronic HG-induced injury through modulation of autophagy in vitro and in vivo. This study unveiled novel evidence of BM-MSCs as an ideal strategy to enhance autophagy for treatment of T2D mellitus.
Collapse
|
121
|
Vaithilingam V, Evans MDM, Rowe A, Bean PA, Tuch BE. Coencapsulation of Target Effector Cells With Mesenchymal Stem Cells Reduces Pericapsular Fibrosis and Improves Graft Survival in a Xenotransplanted Animal Model. Cell Transplant 2015; 25:1299-317. [PMID: 26351069 DOI: 10.3727/096368915x688975] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Pericapsular fibrotic overgrowth (PFO) is a problem that thwarts full implementation of cellular replacement therapies involving encapsulation in an immunoprotective material, such as for the treatment of diabetes. Mesenchymal stem cells (MSCs) have inherent anti-inflammatory properties. We postulated that coencapsulation of MSCs with the target cells would reduce PFO. A hepatoinsulinoma cell line (HUH7) was used to model human target cells and was coencapsulated with either human or mouse MSCs at different ratios in alginate microcapsules. Viability of encapsulated cells was assessed in vitro and xenografted either intraperitoneally or subcutaneously into C57BL/6 mice. Graft retrieval was performed at 3 weeks posttransplantation and assessed for PFO. Coencapsulation of human MSCs (hMSCs) or mouse MSCs (mMSCs) with HUH7 at different ratios did not alter cell viability in vitro. In vivo data from intraperitoneal infusions showed that PFO for HUH7 cells coencapsulated with hMSCs and mMSCs in a ratio of 1:1 was significantly reduced by ∼30% and ∼35%, respectively, compared to HUH7 encapsulated alone. PFO for HUH7 cells was reduced by ∼51% when the ratio of mMSC/HUH7 was increased to 2:1. Implanting the microcapsules subcutaneously rather than intraperitoneally substantially reduced PFO in all treatment groups, which was most significant in the mMSC/HUH7 2:1 group with a ∼53% reduction in PFO compared with HUH7 alone. Despite the reduced PFO reaction to the individual microcapsules implanted subcutaneously, all microcapsule treatment groups were contained in a vascularized fibrotic pouch at 3 weeks. The presence of MSCs in microcapsules retrieved from these fibrotic pouches improved graft survival with significantly higher cell viabilities of 83.1 ± 0.6% and 79.1 ± 0.8% seen with microcapsules containing mMSC/HUH7 at 2:1 and 1:1 ratios, respectively, compared to HUH7 alone (51.5 ± 0.7%) transplanted subcutaneously. This study showed that coencapsulation of MSCs with target cells has a dose-dependent effect on reducing PFO and improving graft survival when implanted either intraperitoneally or subcutaneously in a stringent xenotransplantation setting.
Collapse
|
122
|
Kim KS, Park JM, Kong T, Kim C, Bae SH, Kim HW, Moon J. Retinal Angiogenesis Effects of TGF-β1 and Paracrine Factors Secreted From Human Placental Stem Cells in Response to a Pathological Environment. Cell Transplant 2015; 25:1145-57. [PMID: 26065854 DOI: 10.3727/096368915x688263] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Abnormal angiogenesis is a primary cause of many eye diseases, including diabetic retinopathy, age-related macular degeneration, and retinopathy of prematurity. Mesenchymal stem cells (MSCs) are currently being investigated as a treatment for several such retinal diseases based on their neuroprotective and angiogenic potentials. In this study, we evaluated the role of systemically injected human placental amniotic membrane-derived MSCs (AMSCs) on pathological neovascularization of proliferative retinopathy. We determined that AMSCs secrete higher levels of transforming growth factor-β (TGF-β1) than other MSCs, and the secreted TGF-β1 directly suppresses the proliferation of endothelial cells under pathological conditions in vitro. Moreover, in a mouse model of oxygen-induced retinopathy, intraperitoneally injected AMSCs migrated into the retina and suppressed excessive neovascularization of the vasculature via expression of TGF-β1, and the antineovascular effect of AMSCs was blocked by treatment with TGF-β1 siRNA. These findings are the first to demonstrate that TGF-β1 secreted from AMSCs is one of the key factors to suppress retinal neovascularization in proliferative retinopathy and further elucidate the therapeutic function of AMSCs for the treatment of retinal neovascular diseases.
Collapse
Affiliation(s)
- Kyung-Sul Kim
- College of Life Science, Department of Biotechnology, CHA University, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
123
|
Abuelmagd MM, Nagib AM, Abuelmagd MM, Refaie AF, Elhindi YA, Ahmed MF, Ali MH, Elmaghrabi HM, Bakr MA. Study of the risk factors and complications of diabetes mellitus after live kidney donation. Transplant Proc 2015; 47:1152-7. [PMID: 26036542 DOI: 10.1016/j.transproceed.2014.11.064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 11/13/2014] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Kidney donors, similar to the general population, are at risk for developing type 2 diabetes mellitus (T2DM). The course of donors who develop T2DM has not been well studied. This work estimates the incidence of diabetes after kidney donation, and some risk factors and complications of diabetes mellitus postdonation. MATERIALS AND METHODS This study examined the records of 2267 donors who donated one of their kidneys between 1976 and 2014 at the Urology and Nephrology Center, Mansoura University, Egypt, and who were regularly followed up at its outpatient clinic. A total of 388 donors were included in the study, and their medical records were revised. RESULTS Postdonation weight gain and family history of diabetes mellitus were statistically significant for the development of diabetes mellitus, high or very high albuminuria, and/or decreased creatinine clearance. Metformin and insulin use seemed to significantly reduce the protein excretion and creatinine clearance decline in the studied group. CONCLUSIONS There is a significant impact of a family history of diabetes mellitus on the development of high or very high albuminuria and/or decreased creatinine clearance.
Collapse
Affiliation(s)
- Mohammed M Abuelmagd
- Department of Dialysis and Transplantation, the Urology and Nephrology Center, Mansoura University, Mansoura, Egypt.
| | - A M Nagib
- Department of Dialysis and Transplantation, the Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Megahed M Abuelmagd
- Department of Dialysis and Transplantation, the Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - A F Refaie
- Department of Dialysis and Transplantation, the Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Y A Elhindi
- Department of Dialysis and Transplantation, the Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - M F Ahmed
- Department of Dialysis and Transplantation, the Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - M H Ali
- Department of Dialysis and Transplantation, the Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - H M Elmaghrabi
- Department of Dialysis and Transplantation, the Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - M A Bakr
- Department of Dialysis and Transplantation, the Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| |
Collapse
|
124
|
Bueno PDG, Yochite JNU, Derigge-Pisani GF, Malmegrim de Farias KCR, de Avó LRDS, Voltarelli JC, Leal ÂMDO. Metabolic and pancreatic effects of bone marrow mesenchymal stem cells transplantation in mice fed high-fat diet. PLoS One 2015; 10:e0124369. [PMID: 25923733 PMCID: PMC4414281 DOI: 10.1371/journal.pone.0124369] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/27/2015] [Indexed: 12/22/2022] Open
Abstract
The purpose of this study was to investigate the effects of multiple infusions of allogeneic MSCs on glucose homeostasis and morphometry of pancreatic islets in high- fat diet (HFD) fed mice. Swiss mice were fed standard diet (C group) or HFD (HFD group). After 8 weeks, animals of HFD group received sterile phosphate-buffered saline infusions (HFD-PBS) or four infusions of MSCs one week apart (HFD-MSCs). Fasting glycemia (FG) was determined weekly and glucose (GTT) and insulin (ITT) tolerance tests were performed 4, 8, 12, and 16 weeks after the infusions of MSCs. The MSCs transplanted mice were classified as responder (FG < 180 mg/dL, 72.2% of transplanted mice) or non-responder (FG > 180mg/dL, 28.8%) Seven weeks after MSCs infusions, FG decreased in HFD-MSCs responder mice compared with the HFD-PBS group. Sixteen weeks post MSCs infusions, GTT and ITT areas under the curve (AUC) decreased in HFD-MSCs responder mice compared to HFD-PBS group. Serum insulin concentration was higher in HFD-PBS group than in control animals and was not different compared with the other groups. The relative volume of α-cells was significantly smaller in HFD-PBS group than in C group and significantly higher in HFD-MSCs-NR than in HFD-PBS and HFD-MSCs-R groups. Cell apoptosis in the islets was higher in HFD-PBS group than in C group, and lower in HFD-MSCs responder mice than in HFD-PBS group and non-responder animals. The results demonstrate the ability of multiple infusions of MSCs to promote prolonged decrease in hyperglycemia and apoptosis in pancreatic islets and increase in insulin sensitivity in HFD fed mice.
Collapse
Affiliation(s)
- Patricia de Godoy Bueno
- Department of Physiological Science, Center of Biological Sciences and Health, Federal University of São Carlos, São Carlos, São Paulo, Brazil
| | - Juliana Navarro Ueda Yochite
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Graziela Fernanda Derigge-Pisani
- Department of Physiological Science, Center of Biological Sciences and Health, Federal University of São Carlos, São Carlos, São Paulo, Brazil
| | - Kelen Cristina Ribeiro Malmegrim de Farias
- Department of Clinical, Toxicological and Bromatological Analyses, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirao Preto, São Paulo, Brazil
| | | | | | | |
Collapse
|
125
|
Liu X, Li X, Zhang N, Wen X. Engineering β-cell islets or islet-like structures for type 1 diabetes treatment. Med Hypotheses 2015; 85:82-4. [PMID: 25892491 DOI: 10.1016/j.mehy.2015.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 03/31/2015] [Accepted: 04/05/2015] [Indexed: 01/28/2023]
Abstract
Type 1 diabetes mellitus is a disease characterized by the destruction of the β-cells in the pancreatic islets of Langerhans. The current primary treatment for type 1 diabetes is insulin injections administered multiple times throughout the day. However, this treatment cannot provide sustained physiological release of insulin and the insulin amount is not finely tuned to the glycemia condition. Pancreatic transplantation or islet transplantation would be the preferred treatment strategy but the lack of donor tissue and immunoincompatibility has been shown to be a roadblock to their widespread use. Bioengineering strategies are poised to combat these challenges. Islet encapsulation is expected to offer both immunoisolation and immunomodulation effects by: (1) physically protecting islets from the attacks of immunoglobulins, complements, and host immune cells, and (2) delivering immune regulatory and immunomodulatory factors locally to the islets to protect those islets from immune rejection. Semi-permeable coatings using biocompatible biomaterials can be used for immunoisolating islets away from the host immune systems. Immunoisolation technology also provides an opportunity to use other cell sources for cell therapy to treat type 1 diabetes. Recently, some studies reported that co-transplantation of islets with mesenchymal stem cells (MSCs) can control graft inflammation. MSCs have immunomodulatory property. They are able to secrete anti-inflammatory factors and repress the activity of various immune cells. Growth factors like interleukin 10 (IL-10) and leukemia inhibitory factor (LIF) also have immune regulatory properties. Therefore immunoisolation and immunomodulation technologies can be integrated and applied to β-cell encapsulation for the treatment of type 1 diabetes. Through engineering β-cell islets or islet-like microtissues, the viability and function of transplanted β-cells may be improved. In the meantime, the survival of transplanted β-cells can be further improved by promoting vascular network formation surrounding the transplanted islets or microtissues.
Collapse
Affiliation(s)
| | - Xiaowei Li
- Translational Tissue Engineering Center, Whitaker Biomedical Engineering Institute, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Ning Zhang
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Xuejun Wen
- Institute for Engineering and Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA.
| |
Collapse
|
126
|
EXP CLIN TRANSPLANTExp Clin Transplant 2015; 13. [DOI: 10.6002/ect.mesot2014.o60] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
127
|
Leijten J, Chai Y, Papantoniou I, Geris L, Schrooten J, Luyten F. Cell based advanced therapeutic medicinal products for bone repair: Keep it simple? Adv Drug Deliv Rev 2015; 84:30-44. [PMID: 25451134 DOI: 10.1016/j.addr.2014.10.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 09/18/2014] [Accepted: 10/20/2014] [Indexed: 02/08/2023]
Abstract
The development of cell based advanced therapeutic medicinal products (ATMPs) for bone repair has been expected to revolutionize the health care system for the clinical treatment of bone defects. Despite this great promise, the clinical outcomes of the few cell based ATMPs that have been translated into clinical treatments have been far from impressive. In part, the clinical outcomes have been hampered because of the simplicity of the first wave of products. In response the field has set-out and amassed a plethora of complexities to alleviate the simplicity induced limitations. Many of these potential second wave products have remained "stuck" in the development pipeline. This is due to a number of reasons including the lack of a regulatory framework that has been evolving in the last years and the shortage of enabling technologies for industrial manufacturing to deal with these novel complexities. In this review, we reflect on the current ATMPs and give special attention to novel approaches that are able to provide complexity to ATMPs in a straightforward manner. Moreover, we discuss the potential tools able to produce or predict 'goldilocks' ATMPs, which are neither too simple nor too complex.
Collapse
|
128
|
Atashi F, Modarressi A, Pepper MS. The role of reactive oxygen species in mesenchymal stem cell adipogenic and osteogenic differentiation: a review. Stem Cells Dev 2015; 24:1150-63. [PMID: 25603196 PMCID: PMC4424969 DOI: 10.1089/scd.2014.0484] [Citation(s) in RCA: 486] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are promising candidates for tissue engineering and regenerative medicine. The multipotent stem cell component of MSC isolates is able to differentiate into derivatives of the mesodermal lineage including adipocytes, osteocytes, chondrocytes, and myocytes. Many common pathways have been described in the regulation of adipogenesis and osteogenesis. However, stimulation of osteogenesis appears to suppress adipogenesis and vice-versa. Increasing evidence implicates a tight regulation of these processes by reactive oxygen species (ROS). ROS are short-lived oxygen-containing molecules that display high chemical reactivity toward DNA, RNA, proteins, and lipids. Mitochondrial complexes I and III, and the NADPH oxidase isoform NOX4 are major sources of ROS production during MSC differentiation. ROS are thought to interact with several pathways that affect the transcription machinery required for MSC differentiation including the Wnt, Hedgehog, and FOXO signaling cascades. On the other hand, elevated levels of ROS, defined as oxidative stress, lead to arrest of the MSC cell cycle and apoptosis. Tightly regulated levels of ROS are therefore critical for MSC terminal differentiation, although the precise sources, localization, levels and the exact species of ROS implicated remain to be determined. This review provides a detailed overview of the influence of ROS on adipogenic and osteogenic differentiation in MSCs.
Collapse
Affiliation(s)
- Fatemeh Atashi
- 1 Department of Plastic, Reconstructive & Aesthetic Surgery, University Hospitals of Geneva , University of Geneva, Geneva, Switzerland
| | | | | |
Collapse
|
129
|
Lin HP, Chan TM, Fu RH, Chuu CP, Chiu SC, Tseng YH, Liu SP, Lai KC, Shih MC, Lin ZS, Chen HS, Yeh DC, Lin SZ. Applicability of Adipose-Derived Stem Cells in Type 1 Diabetes Mellitus. Cell Transplant 2015; 24:521-32. [PMID: 25621468 DOI: 10.3727/096368915x686977] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a form of early onset diabetes mellitus characterized by the autoimmune destruction of insulin-producing cells (IPCs), resulting in hyperglycemia and abnormal glucose metabolism. There are currently no treatments available capable of completely curing the symptoms associated with the loss or functional defects of IPCs. Nonetheless, stem cell therapy has demonstrated considerable promise in the replacement of IPCs with immunomodulatory functions to overcome the defects caused by T1DM. Adipose-derived stem cells (ADSCs) are particularly suitable for use in cell transplantation therapy, especially when seeking to avoid the ethical issues and tumorigenic complications commonly associated with embryos or induced pluripotent stem cells. Cell-based treatments have demonstrated therapeutic advantages and clinical applicability of ADSCs in T1DM, ensuring their suitability for transplantation therapy. This manuscript focuses on the benefits and possible mechanisms in a T1DM-relevant model and displays positive results from finished or ongoing human clinical trials. We also discuss and hypothesize potential methods to further enhance the therapeutic efficacy of these efforts, such as a humanized rodent model and gene therapies for IPC clusters, to meet the clinical applicability of the standard.
Collapse
Affiliation(s)
- Hui-Ping Lin
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Tzu-Min Chan
- Department of Medical Education and Research, China Medical University Beigan Hospital, Yunlin, Taiwan
- Department of Medical Education and Research, China Medical University-An-Nan Hospital, Tainan, Taiwan
| | - Ru-Huei Fu
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
| | - Chih-Pin Chuu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Shao-Chih Chiu
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
| | - Yu-Hsiung Tseng
- Department of Nephrology, China Medical University Beigan Hospital, Yunlin, Taiwan
| | - Shih-Ping Liu
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Department of Social Work, Asia University, Taichung, Taiwan
| | - Kuang-Chi Lai
- Department of Surgery, China Medical University Beigan Hospital, Yunlin, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Mu-Chin Shih
- Department of Clinical Laboratory, China Medical University-An-Nan Hospital, Tainan, Taiwan
| | - Zung-Sheng Lin
- Department of Surgery, China Medical University Beigan Hospital, Yunlin, Taiwan
| | - Hsin-Shui Chen
- School of Medicine, China Medical University, Taichung, Taiwan
- Department of Physical Medicine and Rehabilitation, China Medical University Beigang Hospital, Yunlin, Taiwan
| | - Da-Chuan Yeh
- Department of Internal Medicine, China Medical University Beigan Hospital, Yunlin, Taiwan
| | - Shinn-Zong Lin
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
- Department of Neurosurgery, China Medical University Beigan Hospital, Yunlin, Taiwan
- Department of Neurosurgery, China Medical University-An-Nan Hospital, Tainan, Taiwan
| |
Collapse
|
130
|
Yoshimatsu G, Sakata N, Tsuchiya H, Minowa T, Takemura T, Morita H, Hata T, Fukase M, Aoki T, Ishida M, Motoi F, Naitoh T, Katayose Y, Egawa S, Unno M. The co-transplantation of bone marrow derived mesenchymal stem cells reduced inflammation in intramuscular islet transplantation. PLoS One 2015; 10:e0117561. [PMID: 25679812 PMCID: PMC4332659 DOI: 10.1371/journal.pone.0117561] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 12/27/2014] [Indexed: 12/12/2022] Open
Abstract
Aims/Hypothesis Although the muscle is one of the preferable transplant sites in islet transplantation, its transplant efficacy is poor. Here we attempted to determine whether an intramuscular co-transplantation of mesenchymal stem cells (MSCs) could improve the outcome. Methods We co-cultured murine islets with MSCs and then analyzed the morphological changes, viability, insulin-releasing function (represented by the stimulation index), and gene expression of the islets. We also transplanted 500 islets intramuscularly with or without 5 × 105 MSCs to diabetic mice and measured their blood glucose level, the glucose changes in an intraperitoneal glucose tolerance test, and the plasma IL-6 level. Inflammation, apoptosis, and neovascularization in the transplantation site were evaluated histologically. Results The destruction of islets tended to be prevented by co-culture with MSCs. The stimulation index was significantly higher in islets co-cultured with MSCs (1.78 ± 0.59 vs. 7.08 ± 2.53; p = 0.0025). In terms of gene expression, Sult1c2, Gstm1, and Rab37 were significantly upregulated in islets co-cultured with MSCs. Although MSCs were effective in the in vitro assays, they were only partially effective in facilitating intramuscular islet transplantation. Co-transplanted MSCs prevented an early inflammatory reaction from the islets (plasma IL-6; p = 0.0002, neutrophil infiltration; p = 0.016 inflammatory area; p = 0.021), but could not promote neovascularization in the muscle, resulting in the failure of many intramuscular transplanted islets to engraft. Conclusions In conclusion, co-culturing and co-transplanting MSCs is potentially useful in islet transplantation, especially in terms of anti-inflammation, but further augmentation for an anti-apoptosis effect and neovascularization is necessary.
Collapse
Affiliation(s)
- Gumpei Yoshimatsu
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naoaki Sakata
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Haruyuki Tsuchiya
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Minowa
- Nanotechnology Innovation Station, National Institute for Materials Science, Tsukuba, Japan
| | - Taro Takemura
- Nanotechnology Innovation Station, National Institute for Materials Science, Tsukuba, Japan
| | - Hiromi Morita
- Nanotechnology Innovation Station, National Institute for Materials Science, Tsukuba, Japan
| | - Tatsuo Hata
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masahiko Fukase
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takeshi Aoki
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masaharu Ishida
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Fuyuhiko Motoi
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takeshi Naitoh
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yu Katayose
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan; Division of Integrated Surgery and Oncology, Tohoku University Hospital, Sendai, Japan
| | - Shinichi Egawa
- Division of International Cooperation for Disaster Medicine, Tohoku University, Sendai, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
131
|
Shin JY, Jeong JH, Han J, Bhang SH, Jeong GJ, Haque MR, Al-Hilal TA, Noh M, Byun Y, Kim BS. Transplantation of heterospheroids of islet cells and mesenchymal stem cells for effective angiogenesis and antiapoptosis. Tissue Eng Part A 2015; 21:1024-35. [PMID: 25344077 DOI: 10.1089/ten.tea.2014.0022] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Although islet transplantation has been suggested as an alternative therapy for type 1 diabetes, there are efficiency concerns that are attributed to poor engraftment of transplanted islets. Hypoxic condition and delayed vasculogenesis induce necrosis and apoptosis of the transplanted islets. To overcome these limitations in islet transplantation, heterospheroids (HSs), which consist of rat islet cells (ICs) and human bone marrow-derived mesenchymal stem cells (hMSCs), were transplanted to the kidney and liver. The HSs cultured under the hypoxic condition system exhibited a significant increase in antiapoptotic gene expression in ICs. hMSCs in the HSs secreted angiogenic and antiapoptotic proteins. With the HS system, ICs and hMSCs were successfully located in the same area of the liver after transplantation of HSs through the portal vein, whereas the transplantation of islets and the dissociated hMSCs did not result in localization of transplanted ICs and hMSCs in the same area. HS transplantation resulted in an increase in angiogenesis at the transplantation area and a decrease in the apoptosis of transplanted ICs after transplantation into the kidney subcapsule compared with transplantation of islet cell clusters (ICCs). Insulin production levels of ICs were higher in the HS transplantation group compared with the ICC transplantation group. The HS system may be a more efficient transplantation method than the conventional methods for the treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Jung-Youn Shin
- 1 School of Chemical and Biological Engineering, Seoul National University , Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Lee S, Choi E, Cha MJ, Hwang KC. Cell adhesion and long-term survival of transplanted mesenchymal stem cells: a prerequisite for cell therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:632902. [PMID: 25722795 PMCID: PMC4333334 DOI: 10.1155/2015/632902] [Citation(s) in RCA: 189] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 01/19/2015] [Indexed: 12/13/2022]
Abstract
The literature provides abundant evidence that mesenchymal stem cells (MSCs) are an attractive resource for therapeutics and have beneficial effects in regenerating injured tissues due to their self-renewal ability and broad differentiation potential. Although the therapeutic potential of MSCs has been proven in both preclinical and clinical studies, several questions have not yet been addressed. A major limitation to the use of MSCs in clinical applications is their poor viability at the site of injury due to the harsh microenvironment and to anoikis driven by the loss of cell adhesion. To improve the survival of the transplanted MSCs, strategies to regulate apoptotic signaling and enhance cell adhesion have been developed, such as pretreatment with cytokines, growth factors, and antiapoptotic molecules, genetic modifications, and hypoxic preconditioning. More appropriate animal models and a greater understanding of the therapeutic mechanisms of MSCs will be required for their successful clinical application. Nevertheless, the development of stem cell therapies using MSCs has the potential to treat degenerative diseases. This review discusses various approaches to improving MSC survival by inhibiting anoikis.
Collapse
Affiliation(s)
- Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 210-701, Republic of Korea
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City 404-834, Republic of Korea
| | - Eunhyun Choi
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 210-701, Republic of Korea
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City 404-834, Republic of Korea
| | - Min-Ji Cha
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 210-701, Republic of Korea
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City 404-834, Republic of Korea
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 210-701, Republic of Korea
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City 404-834, Republic of Korea
| |
Collapse
|
133
|
Abstract
Apoptosis is one of the major factors contributing to the failure of human islet transplantation. Contributors to islet apoptosis exist in both the pre-transplantation and post transplantation stages. Factors include the islet isolation process, deterioration in vitro prior to transplantation, and immune rejection post transplantation. Previous studies have demonstrated that co-cultured bone marrow cells with human islets not only significantly enhanced the longevity of human islets but also maintained function. We hypothesized that the protective effects of bone marrow cells on human islets are through mechanisms related to preventing apoptosis. This study observed the levels of inflammatory factors such as interleukin-1β (IL-1β), the release of extracellular ATP in vitro, and expression levels of P2X7 ATP receptor (P2X7R), all of which lead to the occurrence of apoptosis in human islets. When human islets were co-cultured with human bone marrow, there was a reduction in the rate of apoptosis correlated with the reduction in inflammatory factors, extra cellular ATP accumulation, and ATP receptor P2X7R expression versus human islets cultured alone. These results suggest that co-culturing bone marrow cells with human islets inhibits inflammation and reduces apoptosis, thus protecting islets from self-deterioration.
Collapse
Affiliation(s)
- Lu-Guang Luo
- Roger Williams Medical Center, Boston University, USA
| | - John Zq Luo
- Brown University, Alpert Medical School, Providence, Rhode Island, USA
| |
Collapse
|
134
|
Hashemian SJ, Kouhnavard M, Nasli-Esfahani E. Mesenchymal Stem Cells: Rising Concerns over Their Application in Treatment of Type One Diabetes Mellitus. J Diabetes Res 2015; 2015:675103. [PMID: 26576437 PMCID: PMC4630398 DOI: 10.1155/2015/675103] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 01/17/2015] [Accepted: 01/18/2015] [Indexed: 12/15/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disorder that leads to beta cell destruction and lowered insulin production. In recent years, stem cell therapies have opened up new horizons to treatment of diabetes mellitus. Among all kinds of stem cells, mesenchymal stem cells (MSCs) have been shown to be an interesting therapeutic option based on their immunomodulatory properties and differentiation potentials confirmed in various experimental and clinical trial studies. In this review, we discuss MSCs differential potentials in differentiation into insulin-producing cells (IPCs) from various sources and also have an overview on currently understood mechanisms through which MSCs exhibit their immunomodulatory effects. Other important issues that are provided in this review, due to their importance in the field of cell therapy, are genetic manipulations (as a new biotechnological method), routes of transplantation, combination of MSCs with other cell types, frequency of transplantation, and special considerations regarding diabetic patients' autologous MSCs transplantation. At the end, utilization of biomaterials either as encapsulation tools or as scaffolds to prevent immune rejection, preparation of tridimensional vascularized microenvironment, and completed or ongoing clinical trials using MSCs are discussed. Despite all unresolved concerns about clinical applications of MSCs, this group of stem cells still remains a promising therapeutic modality for treatment of diabetes.
Collapse
Affiliation(s)
- Seyed Jafar Hashemian
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- *Seyed Jafar Hashemian:
| | - Marjan Kouhnavard
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ensieh Nasli-Esfahani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
135
|
Qi G, Tang Q, Rong R. Dynamic change of glomerular filtration rate in the early stage is associated with kidney allograft status: a preliminary report. Eur J Med Res 2014; 19:72. [PMID: 25539743 PMCID: PMC4297441 DOI: 10.1186/s40001-014-0072-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 12/01/2014] [Indexed: 11/10/2022] Open
Abstract
Background This study aimed to investigate the relationship between the dynamic changes of estimated glomerular filtration rate (eGFR) in the early stage post renal transplantation and renal allograft dysfunction. Methods We selected 9 patients with interstitial fibrosis and tubular atrophy (IF/TA) and 11 patients with stable renal function based on the Banff 2007 classification system. Pathology of the patients was evidenced with renal biopsy results. Glomerular filtration rate (GFR) was calculated continuously for 14 days post-transplantation by using an estimated GFR (eGFR) formula adjusted into Chinese. Linear regression was employed, and eGFR slopes were compared. Prisoners or organs from prisoners were not used in this study. Results and Conclusion The eGFR slope in the IF/TA group was significantly higher than that in the stable group (P < 0.01), and a cut-off value of 5.11 mL/min/1.73 m2/d was a reliable clinical value in a receiver operating characteristic (ROC) curve. On the basis of the ROC area under the curve, predictive accuracy of the eGFR slope was excellent (0.848). In conclusion, the eGFR in IF/TA increased faster within a period of 14 days post-transplantation, suggesting that reperfusion in the early stage may damage the glomerular filtration membrane to some extent. Furthermore, reperfusion might adversely affect long-term renal allograft survival.
Collapse
Affiliation(s)
- Guisheng Qi
- Department of Urology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China. .,Shanghai Key Laboratory of Organ Transplantation, Shanghai, People's Republic of China.
| | - Qunye Tang
- Department of Urology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China. .,Shanghai Key Laboratory of Organ Transplantation, Shanghai, People's Republic of China.
| | - Ruiming Rong
- Department of Urology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China. .,Shanghai Key Laboratory of Organ Transplantation, Shanghai, People's Republic of China. .,Department of Transfusion, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
136
|
Beuscart JB, Pagniez D, Boulanger E, Duhamel A. Registration on the renal transplantation waiting list and mortality on dialysis: an analysis of the French REIN registry using a multi-state model. J Epidemiol 2014; 25:133-41. [PMID: 25721069 PMCID: PMC4310874 DOI: 10.2188/jea.je20130193] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Access to the renal transplantation (RT) waiting list depends on factors related to lower mortality rates and often occurs after dialysis initiation. The aim of the study was to use a flexible regression model to determine if registration on the RT waiting list is associated with mortality on dialysis, independent of the comorbidities associated with such registration. METHODS Data from the French REIN registry on 7138 incident hemodialysis (HD) patients were analyzed. A multi-state model including four states ('HD, not wait-listed', 'HD, wait-listed', 'death', and 'RT') was used to estimate the effect of being wait-listed on the probability of death. RESULTS During the study, 1392 (19.5%) patients were wait-listed. Of the 2954 deaths observed in the entire cohort during follow-up, 2921 (98.9%) were observed in the not wait-listed group compared with only 33 (1.1%) in the wait-listed group. In the multivariable analysis, the adjusted hazard ratio for death associated with non-registration on the waiting list was 3.52 (95% CI, 1.70-7.30). The risk factors for death identified for not wait-listed patients were not found to be significant risk factors for wait-listed patients, with the exception of age. CONCLUSIONS The use of a multi-state model allowed a flexible analysis of mortality on dialysis. Patients who were not wait-listed had a much higher risk of death, regardless of co-morbidities associated with being wait-listed, and did not share the same risk factors of death as wait-listed patients. Registration on the waiting list should therefore be taken into account in survival analysis of patients on dialysis.
Collapse
|
137
|
Bal T, Nazli C, Okcu A, Duruksu G, Karaöz E, Kizilel S. Mesenchymal stem cells and ligand incorporation in biomimetic poly(ethylene glycol) hydrogels significantly improve insulin secretion from pancreatic islets. J Tissue Eng Regen Med 2014; 11:694-703. [PMID: 25393526 DOI: 10.1002/term.1965] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 07/07/2014] [Accepted: 09/25/2014] [Indexed: 12/20/2022]
Abstract
The main goal of this study was to investigate pancreatic islet function with mesenchymal stem cells (MSCs) in a ligand-functionalized poly(ethylene glycol) (PEG) hydrogel for the treatment of type 1 diabetes (T1D). Rat bone marrow-derived MSCs (rBM-MSCs) were encapsulated within synthetic PEG hydrogel, and cell viability and apoptosis within this 3D environment was examined in detail. ATP content and caspase-3 activity of encapsulated MSCs showed that fibronectin-derived RGDS, laminin-derived IKVAV and/or insulinotropic glucagon-like peptide (GLP-1) were required to maintain MSC survival. Incorporation of these peptides into the hydrogel environment also improved pancreatic islet viability, where combinations of peptides had altered effects on islet survival. GLP-1 alone was the leading stimulator for insulin secretion. Cell adhesion peptides RGDS and IKVAV improved insulin secretion only when they were used in combination, but could not surpass the effect of GLP-1. Further, when pancreatic islets were co-encapsulated with MSCs within synthetic PEG hydrogel, a two-fold increase in the stimulation index was measured. Synergistic effects of MSCs and peptides were observed, with a seven-fold increase in the stimulation index. The results are promising and suggest that simultaneous incorporation of MSCs and ECM-derived peptides and/or GLP-1 can improve pancreatic islet function in response to altered glucose levels in the physiological environment. Copyright © 2014 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Tuğba Bal
- Chemical and Biological Engineering, Koc University, Istanbul, Turkey
| | - Caner Nazli
- Material Sciences and Engineering, Koc University, Istanbul, Turkey
| | - Alparslan Okcu
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey
| | - Gökhan Duruksu
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey
| | - Erdal Karaöz
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey.,Liv Hospital, Regenerative Medicine, Stem Cell Research and Therapy Center, Istanbul, Turkey
| | - Seda Kizilel
- Chemical and Biological Engineering, Koc University, Istanbul, Turkey.,Material Sciences and Engineering, Koc University, Istanbul, Turkey
| |
Collapse
|
138
|
Kosmoliaptsis V, Gjorgjimajkoska O, Sharples LD, Chaudhry AN, Chatzizacharias N, Peacock S, Torpey N, Bolton EM, Taylor CJ, Bradley JA. Impact of donor mismatches at individual HLA-A, -B, -C, -DR, and -DQ loci on the development of HLA-specific antibodies in patients listed for repeat renal transplantation. Kidney Int 2014; 86:1039-1048. [PMID: 24717292 DOI: 10.1038/ki.2014.106] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 01/31/2014] [Accepted: 02/13/2014] [Indexed: 12/22/2022]
Abstract
We have analyzed the relationship between donor mismatches at each HLA locus and development of HLA locus-specific antibodies in patients listed for repeat transplantation. HLA antibody screening was undertaken using single-antigen beads in 131 kidney transplant recipients returning to the transplant waiting list following first graft failure. The number of HLA mismatches and the calculated reaction frequency of antibody reactivity against 10,000 consecutive deceased organ donors were determined for each HLA locus. Two-thirds of patients awaiting repeat transplantation were sensitized (calculated reaction frequency over 15%) and half were highly sensitized (calculated reaction frequency of 85% and greater). Antibody levels peaked after re-listing for repeat transplantation, were independent of graft nephrectomy and were associated with length of time on the waiting list (odds ratio 8.4) and with maintenance on dual immunosuppression (odds ratio 0.2). Sensitization was independently associated with increasing number of donor HLA mismatches (odds ratio 1.4). All mismatched HLA loci contributed to the development of HLA locus-specific antibodies (HLA-A: odds ratio 3.2, HLA-B: odds ratio 3.4, HLA-C: odds ratio 2.5, HLA-DRB1: odds ratio 3.5, HLA-DRB3/4/5: odds ratio 3.9, and HLA-DQ: odds ratio 3.0 (all significant)). Thus, the risk of allosensitization following failure of a first renal transplant increases incrementally with the number of mismatches at all HLA loci assessed. Maintenance of re-listed patients on dual immunosuppression was associated with a reduced risk of sensitization.
Collapse
Affiliation(s)
- Vasilios Kosmoliaptsis
- Department of Surgery, University of Cambridge, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - Olivera Gjorgjimajkoska
- Department of Surgery, University of Cambridge, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - Linda D Sharples
- MRC Biostatistics Unit, Institute of Public Health, Cambridge, UK
| | - Afzal N Chaudhry
- Department of Renal Medicine, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - Nikolaos Chatzizacharias
- Department of Surgery, University of Cambridge, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - Sarah Peacock
- Histocompatibility and Immunogenetics Laboratory, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - Nicholas Torpey
- Department of Renal Medicine, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - Eleanor M Bolton
- Department of Surgery, University of Cambridge, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - Craig J Taylor
- Histocompatibility and Immunogenetics Laboratory, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - J Andrew Bradley
- Department of Surgery, University of Cambridge, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
139
|
Takahashi T, Tibell A, Ljung K, Saito Y, Gronlund A, Osterholm C, Holgersson J, Lundgren T, Ericzon BG, Corbascio M, Kumagai-Braesch M. Multipotent mesenchymal stromal cells synergize with costimulation blockade in the inhibition of immune responses and the induction of Foxp3+ regulatory T cells. Stem Cells Transl Med 2014; 3:1484-94. [PMID: 25313200 DOI: 10.5966/sctm.2014-0012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Multipotent mesenchymal stromal cell (MSC) therapy and costimulation blockade are two immunomodulatory strategies being developed concomitantly for the treatment of immunological diseases. Both of these strategies have the capacity to inhibit immune responses and induce regulatory T cells; however, their ability to synergize remains largely unexplored. In order to study this, MSCs from C57BL/6 (H2b) mice were infused together with fully major histocompatibility complex-mismatched Balb/c (H2d) allogeneic islets into the portal vein of diabetic C57BL/6 (H2b) mice, which were subsequently treated with costimulation blockade for the first 10 days after transplantation. Mice receiving both recipient-type MSCs, CTLA4Ig, and anti-CD40L demonstrated indefinite graft acceptance, just as did most of the recipients receiving MSCs and CTLA4Ig. Recipients of MSCs only rejected their grafts, and fewer than one half of the recipients treated with costimulation blockade alone achieved permanent engraftment. The livers of the recipients treated with MSCs plus costimulation blockade contained large numbers of islets surrounded by Foxp3+ regulatory T cells. These recipients showed reduced antidonor IgG levels and a glucose tolerance similar to that of naïve nondiabetic mice. Intrahepatic lymphocytes and splenocytes from these recipients displayed reduced proliferation and interferon-γ production when re-exposed to donor antigen. MSCs in the presence of costimulation blockade prevented dendritic cell maturation, inhibited T cell proliferation, increased Foxp3+ regulatory T cell numbers, and increased indoleamine 2,3-dioxygenase activity. These results indicate that MSC infusion and costimulation blockade have complementary immune-modulating effects that can be used for a broad number of applications in transplantation, autoimmunity, and regenerative medicine.
Collapse
Affiliation(s)
- Tohru Takahashi
- Division of Transplantation Surgery, Karolinska Institutet, CLINTEC, Stockholm, Sweden; Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry and Transfusion Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Cardiothoracic Surgery and Anesthesiology, Karolinksa University Hospital, Stockholm, Sweden
| | - Annika Tibell
- Division of Transplantation Surgery, Karolinska Institutet, CLINTEC, Stockholm, Sweden; Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry and Transfusion Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Cardiothoracic Surgery and Anesthesiology, Karolinksa University Hospital, Stockholm, Sweden
| | - Karin Ljung
- Division of Transplantation Surgery, Karolinska Institutet, CLINTEC, Stockholm, Sweden; Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry and Transfusion Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Cardiothoracic Surgery and Anesthesiology, Karolinksa University Hospital, Stockholm, Sweden
| | - Yu Saito
- Division of Transplantation Surgery, Karolinska Institutet, CLINTEC, Stockholm, Sweden; Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry and Transfusion Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Cardiothoracic Surgery and Anesthesiology, Karolinksa University Hospital, Stockholm, Sweden
| | - Anna Gronlund
- Division of Transplantation Surgery, Karolinska Institutet, CLINTEC, Stockholm, Sweden; Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry and Transfusion Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Cardiothoracic Surgery and Anesthesiology, Karolinksa University Hospital, Stockholm, Sweden
| | - Cecilia Osterholm
- Division of Transplantation Surgery, Karolinska Institutet, CLINTEC, Stockholm, Sweden; Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry and Transfusion Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Cardiothoracic Surgery and Anesthesiology, Karolinksa University Hospital, Stockholm, Sweden
| | - Jan Holgersson
- Division of Transplantation Surgery, Karolinska Institutet, CLINTEC, Stockholm, Sweden; Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry and Transfusion Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Cardiothoracic Surgery and Anesthesiology, Karolinksa University Hospital, Stockholm, Sweden
| | - Torbjörn Lundgren
- Division of Transplantation Surgery, Karolinska Institutet, CLINTEC, Stockholm, Sweden; Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry and Transfusion Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Cardiothoracic Surgery and Anesthesiology, Karolinksa University Hospital, Stockholm, Sweden
| | - Bo-Göran Ericzon
- Division of Transplantation Surgery, Karolinska Institutet, CLINTEC, Stockholm, Sweden; Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry and Transfusion Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Cardiothoracic Surgery and Anesthesiology, Karolinksa University Hospital, Stockholm, Sweden
| | - Matthias Corbascio
- Division of Transplantation Surgery, Karolinska Institutet, CLINTEC, Stockholm, Sweden; Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry and Transfusion Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Cardiothoracic Surgery and Anesthesiology, Karolinksa University Hospital, Stockholm, Sweden
| | - Makiko Kumagai-Braesch
- Division of Transplantation Surgery, Karolinska Institutet, CLINTEC, Stockholm, Sweden; Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry and Transfusion Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Cardiothoracic Surgery and Anesthesiology, Karolinksa University Hospital, Stockholm, Sweden
| |
Collapse
|
140
|
Kwon HM, Hur SM, Park KY, Kim CK, Kim YM, Kim HS, Shin HC, Won MH, Ha KS, Kwon YG, Lee DH, Kim YM. Multiple paracrine factors secreted by mesenchymal stem cells contribute to angiogenesis. Vascul Pharmacol 2014; 63:19-28. [DOI: 10.1016/j.vph.2014.06.004] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 06/02/2014] [Accepted: 06/20/2014] [Indexed: 01/08/2023]
|
141
|
Immunogenicity of allogeneic mesenchymal stem cells transplanted via different routes in diabetic rats. Cell Mol Immunol 2014; 12:444-55. [PMID: 25242276 DOI: 10.1038/cmi.2014.70] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 07/02/2014] [Accepted: 07/03/2014] [Indexed: 02/06/2023] Open
Abstract
Due to their hypoimmunogenicity and unique immunosuppressive properties, mesenchymal stem cells (MSCs) are considered one of the most promising adult stem cell types for cell therapy. Although many studies have shown that MSCs exert therapeutic effects on several acute and subacute conditions, their long-term effects are not confirmed in chronic diseases. Immunogenicity is a major limitation for cell replacement therapy, and it is not well understood in vivo. We evaluated the immunogenicity of allogeneic MSCs in vivo by transplanting MSCs into normal and diabetic rats via the tail vein or pancreas and found that MSCs exhibited low immunogenicity in normal recipients and even exerted some immunosuppressive effects in diabetic rats during the initial phase. However, during the later stage in the pancreas group, MSCs expressed insulin and MHC II, eliciting a strong immune response in the pancreas. Simultaneously, the peripheral blood mononuclear cells in the recipients in the pancreas group were activated, and alloantibodies developed in vivo. Conversely, in the tail vein group, MSCs remained immunoprivileged and displayed immunosuppressive effects in vivo. These data indicate that different transplanting routes and microenvironments can lead to divergent immunogenicity of MSCs.
Collapse
|
142
|
Park KS, Shin JH, Jang HR, Lee JE, Huh WS, Kim YG, Oh HY, Kim DJ. Impact of donor kidney function and donor age on poor outcome of living-unrelated kidney transplantation (KT) in comparison with living-related KT. Clin Transplant 2014; 28:953-60. [PMID: 24861232 DOI: 10.1111/ctr.12388] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2014] [Indexed: 12/01/2022]
Abstract
Living-unrelated donors (LURD) have been widely used for kidney transplantation (KT). We retrospectively reviewed 779 patients who underwent living-donor KT from 2000 to 2012, to compare outcomes of 264 KT from LURD and 515 from living-related donors (LRD), and to identify risk factors for living KT. Median follow-up was 67 months. Mean donor age, total human leukocyte antigen (HLA) mismatches, and HLA-DR mismatches were higher, and mean estimated glomerular filtration rate (eGFR) was lower in LURD. Acute rejection (AR)-free survival (p = 0.018) and graft survival (p = 0.025) were lower for LURD than LRD, whereas patient survival rate was comparable. Cox regression analysis showed HLA-DR mismatches (OR 1.75 for one mismatch; OR 2.19 for two mismatches), recipient age ≤ 42 yr, and donor age > 50 yr were significant risk factors for acute rejection. For graft survival, AR and donor eGFR (OR 1.90, p = 0.035) were significant. We also identified significant impact of recipient age > 50 yr and diabetes for patient survival. However, KT from LURD was not a significant risk factor for AR (p = 0.368), graft survival (p = 0.205), and patient survival (p = 0.836). Our data suggest that donor eGFR and donor age are independent risk factors for clinical outcomes of living KT, which can be related with poor outcome of KT from LURD.
Collapse
Affiliation(s)
- Kyung Sun Park
- Division of Nephrology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
143
|
Ayaz S, Gençoğlu EA, Moray G, Gözükara MY, Haberal M. Evaluation of the effects of recipient/donor gender on early/late postoperative renal graft functions by renal scintigraphy. EXP CLIN TRANSPLANT 2014; 12:510-4. [PMID: 24918693 DOI: 10.6002/ect.2013.0275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES We discuss the effects of recipient/donor gender on renal allograft functions using scintigraphic parameters obtained 3 days after renal transplant and 1 year after transplant. MATERIALS AND METHODS This retrospective study included 76 renal allograft recipients (group one, 38 males; group two, 38 females). Patients underwent scintigraphic imaging with Tc-99m DTPA on postoperative day 3 and 1 year after transplant. We used the Hilson perfusion index, maximum renal activity/background activity, ratio of renal activity at 20 minutes to renal activity at 3 minutes, time-to-peak activity, and glomerular filtration rate to measure quantitative parameters. RESULTS On postoperative day 3, the Hilson perfusion index, maximum renal activity/background activity, the ratio of renal activity at 20 minutes to renal activity at 3 minutes, time-to-peak activity, and glomerular filtration rate values for male/female recipients were similar (P = .65, P = .77, P = .38, P = .10, P = .99). The gender of donors was compared with the above-mentioned scintigraphic parameters of the recipients, and no statistically significant differences were found (P = .24, P = .25, P = .44, P = .29, P = .13). At 1-year follow-up, values obtained from group 1 and group 2 recipients were similar. After 1 year, chronic rejection developed in 15.7% of group 1 recipients and in 10.5% of group 2 recipients; acute rejection developed in 21% of group 1 recipients and in 23.6% of group 2 recipients. There were no statistically significant differences between the occurrence of acute rejection and the gender of recipients or donors (P = 1.00, P = .45). CONCLUSIONS We observed no statistically significant differences between renal graft functions and gender of the recipients/donors during the early/late posttransplant period.
Collapse
Affiliation(s)
- Sevin Ayaz
- From the Department of Nuclear Medicine, Mersin State Hospital, Mersin, Turkey
| | | | | | | | | |
Collapse
|
144
|
Tan C, Lui PPY, Lee YW, Wong YM. Scx-transduced tendon-derived stem cells (tdscs) promoted better tendon repair compared to mock-transduced cells in a rat patellar tendon window injury model. PLoS One 2014; 9:e97453. [PMID: 24831949 PMCID: PMC4022525 DOI: 10.1371/journal.pone.0097453] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 04/21/2014] [Indexed: 02/06/2023] Open
Abstract
We hypothesized that the transplantation of Scx-transduced tendon-derived stem cells (TDSCs) promoted better tendon repair compared to the transplantation of mock-transduced cells. This study thus aimed to investigate the effect of Scx transduction on the expression of lineage markers in TDSCs and the effect of the resulting cell line in the promotion of tendon repair. Rat non-GFP or GFP-TDSCs were transduced with Scx or empty lentiviral vector (Mock) and selected by blasticidin. The mRNA expressions of Scx and different lineage markers were examined by qRT-PCR. The effect of the transplantation of GFP-TDSC-Scx on tendon repair was then tested in a rat unilateral patellar tendon window injury model. The transplantation of GFP-TDSC-Mock and scaffold-only served as controls. At week 2, 4 and 8 post-transplantation, the repaired patellar tendon was harvested for ex vivo fluorescent imaging, vivaCT imaging, histology, immunohistochemistry and biomechanical test. GFP-TDSC-Scx consistently showed higher expressions of most of tendon- and cartilage- related markers compared to the GFP-TDSC-Mock. However, the effect of Scx transduction on the expressions of bone-related markers was inconclusive. The transplanted GFP-TDSCs could be detected in the window wound at week 2 but not at week 4. Ectopic mineralization was detected in some samples at week 8 but there was no difference among different groups. The GFP-TDSC-Scx group only statistically significantly improved tendon repair histologically and biomechanically compared to the Scaffold-only group and the GFP-TDSC-Mock group at the early stage of tendon repair. There was significant higher expression of collagen type I in the window wound in the GFP-TDSC-Scx group compared to the other two groups at week 2. The transplantation of GFP-TDSC-Scx promoted healing at the early stage of tendon repair in a rat patellar tendon window injury model.
Collapse
Affiliation(s)
- Chunlai Tan
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- The Hong Kong Jockey Club Sports Medicine and Health Sciences Centre, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | | | - Yuk Wa Lee
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- The Hong Kong Jockey Club Sports Medicine and Health Sciences Centre, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yin Mei Wong
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- The Hong Kong Jockey Club Sports Medicine and Health Sciences Centre, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
145
|
Gao X, Song L, Shen K, Wang H, Qian M, Niu W, Qin X. Bone marrow mesenchymal stem cells promote the repair of islets from diabetic mice through paracrine actions. Mol Cell Endocrinol 2014; 388:41-50. [PMID: 24667703 DOI: 10.1016/j.mce.2014.03.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 01/18/2014] [Accepted: 03/07/2014] [Indexed: 12/31/2022]
Abstract
Transplantation of bone marrow mesenchymal stem cells (MSCs) has been shown to effectively lower blood glucose levels in diabetic individuals, but the mechanism has not been adequately explained. We hypothesized that MSCs exert beneficial paracrine actions on the injured islets by releasing biologically active factors. To prove our hypothesis, we tested the cytoprotective effect of conditioned medium from cultured MSCs on isolated islets exposed to STZ in vitro and on mice islets after the experimental induction of diabetes in vivo. We assessed islet regeneration in the presence of conditioned medium and explored the possible mechanisms involved. Transplantation of MSCs can ameliorate hyperglycemia in diabetic mice by promoting the regeneration of β cells. Both β cell replication and islet progenitors differentiation contribute to β cell regeneration. MSC transplantation resulted in increases in pAkt and pErk expression by islets in vivo. Treatment with MSC-CM promoted islet cell proliferation and resulted in increases in pAkt and pErk expression by islets in vitro. The MSC-CM-mediated induction of β cell proliferation was completely blocked by the PI3K/Akt inhibitor LY294002 but not by the MEK/Erk inhibitor PD98059. Together, these data suggest that the PI3K/Akt signal pathway plays a critical role in β cell proliferation after MSC transplantation.
Collapse
Affiliation(s)
- Xiaodong Gao
- Department of General Surgery, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Institute of General Surgery, Fudan University, Shanghai, People's Republic of China
| | - Lujun Song
- Department of General Surgery, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Institute of General Surgery, Fudan University, Shanghai, People's Republic of China
| | - Kuntang Shen
- Department of General Surgery, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Institute of General Surgery, Fudan University, Shanghai, People's Republic of China
| | - Hongshan Wang
- Department of General Surgery, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Institute of General Surgery, Fudan University, Shanghai, People's Republic of China
| | - Mengjia Qian
- Experimental Research Center, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Weixin Niu
- Department of General Surgery, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
| | - Xinyu Qin
- Department of General Surgery, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Institute of General Surgery, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
146
|
The efficacy of mesenchymal stem cell transplantation in caustic esophagus injury: an experimental study. Stem Cells Int 2014; 2014:939674. [PMID: 24876849 PMCID: PMC4027018 DOI: 10.1155/2014/939674] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Accepted: 04/07/2014] [Indexed: 12/11/2022] Open
Abstract
Introduction. Ingestion of corrosive substances may lead to stricture formation in esophagus as a late complication. Full thickness injury seems to exterminate tissue stem cells of esophagus. Mesenchymal stem cells (MSCs) can differentiate into specific cell lineages and have the capacity of homing in sites of injury. Aim and Methods. We aimed to investigate the efficacy of MSC transplantation, on prevention of esophageal damage and stricture formation after caustic esophagus injury in rats. 54 rats were allocated into four groups; 4 rats were sacrificed for MSC production. Group 1, untreated controls (n: 10). Group 2, membrane labeled MSCs-treated rats (n: 20). Group 3, biodistribution of fluorodeoxyglucose labeled MSCs via positron emission tomography (PET) imaging (n: 10). Group 4, sham operated (n: 10). Standard caustic esophageal burns were created and MSCs were transplanted 24 hours after. All rats were sacrificed at the 21st days. Results. PET scan images revealed the homing behavior of MSCs to the injury site. The histopathology damage score was not significantly different from controls. However, we demonstrated Dil labeled epithelial and muscle cells which were originating from transplanted MSCs. Conclusion. MSC transplantation after caustic esophageal injury may be a helpful treatment modality; however, probably repeated infusions are needed.
Collapse
|
147
|
Rackham CL, Dhadda PK, Le Lay AM, King AJF, Jones PM. Preculturing Islets With Adipose-Derived Mesenchymal Stromal Cells Is an Effective Strategy for Improving Transplantation Efficiency at the Clinically Preferred Intraportal Site. CELL MEDICINE 2014; 7:37-47. [PMID: 26858891 DOI: 10.3727/215517914x680047] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We have recently shown that preculturing islets with kidney-derived mesenchymal stromal cells (MSCs) improves transplantation outcome in streptozotocin-diabetic mice implanted with a minimal mass of islets beneath the kidney capsule. In the present study, we have extended our previous observations to investigate whether preculturing islets with MSCs can also be used to enhance islet function at the clinically used intraportal site. We have used MSCs derived from adipose tissue, which are more readily accessible than alternative sources in human subjects and can be expanded to clinically efficacious numbers, to preculture islets throughout this study. The in vivo efficacy of grafts consisting of islets precultured alone or with MSCs was tested using a syngeneic streptozotocin-diabetic minimal islet mass model at the clinically relevant intraportal site. Blood glucose concentrations were monitored for 1 month. The vascularization of islets precultured alone or with MSCs was investigated both in vitro and in vivo, using immunohistochemistry. Islet insulin content was measured by radioimmunoassay. The effect of preculturing islets with MSCs on islet function in vitro was investigated using static incubation assays. There was no beneficial angiogenic influence of MSC preculture, as demonstrated by the comparable vascularization of islets precultured alone or with MSCs, both in vitro after 3 days and in vivo 1 month after islet transplantation. However, the in vitro insulin secretory capacity of MSC precultured islets was superior to that of islets precultured alone. In vivo, this was associated with improved glycemia at 7, 14, 21, and 28 days posttransplantation, in recipients of MSC precultured islets compared to islets precultured alone. The area of individual islets within the graft-bearing liver was significantly higher in recipients of MSC precultured islets compared to islets precultured alone. Our experimental studies suggest that preculturing islets with MSCs represents a favorable strategy for improving the efficiency of clinical islet transplantation.
Collapse
Affiliation(s)
- Chloe L Rackham
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, King's College London , London , UK
| | - Paramjeet K Dhadda
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, King's College London , London , UK
| | - Aurélie M Le Lay
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, King's College London , London , UK
| | - Aileen J F King
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, King's College London , London , UK
| | - Peter M Jones
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, King's College London , London , UK
| |
Collapse
|
148
|
Niemi M, Mandelbrot DA. The Outcomes of Living Kidney Donation from Medically Complex Donors: Implications for the Donor and the Recipient. CURRENT TRANSPLANTATION REPORTS 2014; 1:1-9. [PMID: 24579060 PMCID: PMC3933185 DOI: 10.1007/s40472-013-0001-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Living kidney donation is an important option for patients with end-stage renal disease (ESRD), and has improved life expectancy and quality for patients otherwise requiring maintenance dialysis or deceased-donor transplantation. Given the favorable outcomes of live donation and the shortage of organs to transplant, individuals with potentially unfavorable demographic and clinical characteristics are increasingly being permitted to donate kidneys. While this trend has successfully expanded the live donor pool, it has raised concerns as to which acceptance criteria are safe. This review aims to summarize the existing literature on the outcomes of transplantation from medically complex, living kidney donors, including both donor and recipient outcomes when available.
Collapse
Affiliation(s)
- Matthew Niemi
- Division of Nephrology, Department of Medicine Beth Israel Deaconess Medical Center 185 Pilgrim Road, Farr 8 Boston, MA 02215
| | - Didier A Mandelbrot
- The Transplant Center Beth Israel Deaconess Medical Center 110 Francis Street, LMOB 7 Boston, MA 02215
| |
Collapse
|
149
|
Abstract
Cell therapy has enormous potential for the treatment of conditions of unmet medical need. Cell therapy may be applied to diabetes mellitus in the context of beta cell replacement or for the treatment of diabetic complications. A large number of cell types including hematopoietic stem cells, mesenchymal stem cells, umbilical cord blood, conditioned lymphocytes, mononuclear cells, or a combination of these cells have been shown to be safe and feasible for the treatment of patients with diabetes mellitus. The first part of this review article will focus on the current perspective of the role of embryonic stem cells and inducible pluripotent stem cells for beta cell replacement and the current clinical data on cell-based therapy for the restoration of normoglycemia. The second part of this review will highlight the therapeutic role of MSCs in islet cells cotransplantation and the management of diabetes related vascular complications.
Collapse
Affiliation(s)
- Aaron Liew
- Regenerative Medicine Institute (REMEDI), National Centre for Biomedical Engineering Science (NCBES), National University Ireland Galway (NUIG), Galway, Ireland
| | | |
Collapse
|
150
|
Borg DJ, Weigelt M, Wilhelm C, Gerlach M, Bickle M, Speier S, Bonifacio E, Hommel A. Mesenchymal stromal cells improve transplanted islet survival and islet function in a syngeneic mouse model. Diabetologia 2014; 57:522-31. [PMID: 24253203 DOI: 10.1007/s00125-013-3109-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 10/23/2013] [Indexed: 12/13/2022]
Abstract
AIMS/HYPOTHESIS Islet transplantation is used therapeutically in a minority of patients with type 1 diabetes. Successful outcomes are hampered by early islet beta cell loss. The adjuvant co-transplantation of mesenchymal stromal cells (MSCs) has the promise to improve islet transplant outcome. METHODS We used a syngeneic marginal islet mass transplantation model in a mouse model of diabetes. Mice received islets or islets plus 250,000 MSCs. Kidney subcapsule, intra-hepatic and intra-ocular islet transplantation sites were used. Apoptosis, vascularisation, beta cell proliferation, MSC differentiation and laminin levels were determined by immunohistochemical analysis and image quantification post-transplant. RESULTS Glucose homeostasis after the transplantation of syngeneic islets was improved by the co-transplantation of MSCs together with islets under the kidney capsule (p = 0.01) and by intravenous infusion of MSCs after intra-hepatic islet transplantation (p = 0.05). MSC co-transplantation resulted in reduced islet apoptosis, with reduced numbers of islet cells positive for cleaved caspase 3 being observed 14 days post-transplant. In kidney subcapsule, but not in intra-ocular islet transplant models, we observed increased re-vascularisation rates, but not increased blood vessel density in and around islets co-transplanted with MSCs compared with islets that were transplanted alone. Co-transplantation of MSCs did not increase beta cell proliferation, extracellular matrix protein laminin production or alpha cell numbers, and there was negligible MSC transdifferentiation into beta cells. CONCLUSIONS/INTERPRETATION Co-transplantation of MSCs may lead to improved islet function and survival in the early post-transplantation period in humans receiving islet transplantation.
Collapse
Affiliation(s)
- Danielle J Borg
- DFG-Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstrasse 105, 01307, Dresden, Germany
| | | | | | | | | | | | | | | |
Collapse
|