1
|
Siemionow M, Chambily L, Brodowska S. Efficacy of Engraftment and Safety of Human Umbilical Di-Chimeric Cell (HUDC) Therapy after Systemic Intraosseous Administration in an Experimental Model. Biomedicines 2024; 12:1064. [PMID: 38791026 PMCID: PMC11117770 DOI: 10.3390/biomedicines12051064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 04/30/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Cell-based therapies hold promise for novel therapeutic strategies in regenerative medicine. We previously characterized in vitro human umbilical di-chimeric cells (HUDCs) created via the ex vivo fusion of human umbilical cord blood (UCB) cells derived from two unrelated donors. In this in vivo study, we assessed HUDC safety and biodistribution in the NOD SCID mouse model at 90 days following the systemic intraosseous administration of HUDCs. Twelve NOD SCID mice (n = 6/group) received intraosseous injection of donor UCB cells (3.0 × 106) in Group 1, or HUDCs (3.0 × 106) in Group 2, without immunosuppression. Flow cytometry assessed hematopoietic cell surface markers in peripheral blood and the presence of HLA-ABC class I antigens in lymphoid and non-lymphoid organs. HUDC safety was assessed by weekly evaluations, magnetic resonance imaging (MRI), and at autopsy for tumorigenicity. At 90 days after intraosseous cell administration, the comparable expression of HLA-ABC class I antigens in selected organs was found in UCB control and HUDC therapy groups. MRI and autopsy confirmed safety by no signs of tumor growth. This study confirmed HUDC biodistribution to selected lymphoid organs following intraosseous administration, without immunosuppression. These data introduce HUDCs as a novel promising approach for immunomodulation in transplantation.
Collapse
Affiliation(s)
- Maria Siemionow
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL 60607, USA; (L.C.); (S.B.)
- Department of Traumatology, Orthopaedics, and Surgery of the Hand, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Lucile Chambily
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL 60607, USA; (L.C.); (S.B.)
| | - Sonia Brodowska
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL 60607, USA; (L.C.); (S.B.)
| |
Collapse
|
2
|
Narzisi A, Halladay A, Masi G, Novarino G, Lord C. Tempering expectations: considerations on the current state of stem cells therapy for autism treatment. Front Psychiatry 2023; 14:1287879. [PMID: 37854442 PMCID: PMC10579796 DOI: 10.3389/fpsyt.2023.1287879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 09/13/2023] [Indexed: 10/20/2023] Open
Affiliation(s)
- Antonio Narzisi
- Department of Child Psychiatry and Psychopharmacology, IRCCS Stella Maris Foundation, Calambrone, Pisa, Italy
| | - Alycia Halladay
- Autism Science Foundation, New York, NY, United States
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, United States
| | - Gabriele Masi
- Department of Child Psychiatry and Psychopharmacology, IRCCS Stella Maris Foundation, Calambrone, Pisa, Italy
| | - Gaia Novarino
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Catherine Lord
- Department of Psychiatry and Human Development and Psychology, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
3
|
Devi S, Bongale AM, Tefera MA, Dixit P, Bhanap P. Fresh Umbilical Cord Blood-A Source of Multipotent Stem Cells, Collection, Banking, Cryopreservation, and Ethical Concerns. Life (Basel) 2023; 13:1794. [PMID: 37763198 PMCID: PMC10533013 DOI: 10.3390/life13091794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/02/2023] [Accepted: 05/25/2023] [Indexed: 09/29/2023] Open
Abstract
Umbilical cord blood (UCB) is a rich source of hematopoietic cells that can be used to replace bone marrow components. Many blood disorders and systemic illnesses are increasingly being treated with stem cells as regenerative medical therapy. Presently, collected blood has been stored in either public or private banks for allogenic or autologous transplantation. Using a specific keyword, we used the English language to search for relevant articles in SCOPUS and PubMed databases over time frame. According to our review, Asian countries are increasingly using UCB preservation for future use as regenerative medicine, and existing studies indicate that this trend will continue. This recent literature review explains the methodology of UCB collection, banking, and cryopreservation for future clinical use. Between 2010 and 2022, 10,054 UCB stem cell samples were effectively cryopreserved. Furthermore, we have discussed using Mesenchymal Stem Cells (MSCs) as transplant medicine, and its clinical applications. It is essential for healthcare personnel, particularly those working in labor rooms, to comprehend the protocols for collecting, transporting, and storing UCB. This review aims to provide a glimpse of the details about the UCB collection and banking processes, its benefits, and the use of UCB-derived stem cells in clinical practice, as well as the ethical concerns associated with UCB, all of which are important for healthcare professionals, particularly those working in maternity wards; namely, the obstetrician, neonatologist, and anyone involved in perinatal care. This article also highlights the practical and ethical concerns associated with private UCB banks, and the existence of public banks. UCB may continue to grow to assist healthcare teams worldwide in treating various metabolic, hematological, and immunodeficiency disorders.
Collapse
Affiliation(s)
- Seeta Devi
- Department of Obstetrics and Gynecological Nursing, Symbiosis College of Nursing, Symbiosis International (Deemed University), Lavale, Pune 412 115, Maharashtra, India;
| | - Anupkumar M. Bongale
- Department of Artificial Intelligence and Machine Learning, Symbiosis Institute of Technology, Symbiosis International (Deemed University), Lavale, Pune 412 115, Maharashtra, India
| | | | | | - Prasad Bhanap
- HoD OBG Department, Symbiosis Medical College for Women (SMCW), Symbiosis International (Deemed University), Lavale, Pune 412 115, Maharashtra, India
| |
Collapse
|
4
|
Vinukonda G, La Gamma EF. Emerging therapies for brain recovery after IVH in neonates: Cord blood derived Mesenchymal Stem Cells (MSC) and Unrestricted Somatic Stem Cells (USSC). Semin Perinatol 2022; 46:151598. [PMID: 35589461 DOI: 10.1016/j.semperi.2022.151598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this report, we summarize evidence on mechanisms of injury after intraventricular hemorrhage resulting in post-hemorrhagic white matter injury and hydrocephalus and correlate that with the possibility of cellular therapy. We describe how two stem cell lines (MSC & USSC) acting in a paracrine fashion offer promise for attenuating the magnitude of injury in animal models and for improved functional recovery by: lowering the magnitude of apoptosis and neuronal cell death, reducing inflammation, and thus, mitigating white matter injury that culminates in improved motor and neurocognitive outcomes. Animal models of IVH are analyzed for their similarity to the human condition and we discuss merits of each approach. Studies on stem cell therapy for IVH in human neonates is described. Lastly, we offer suggestions on what future studies are needed to better understand mechanisms of injury and recovery and argue that human trials need to be expanded in parallel to animal research.
Collapse
Affiliation(s)
- Govindaiah Vinukonda
- Department of Pediatrics, Cell Biology & Anatomy New York Medical College, Valhalla, NY
| | - Edmund F La Gamma
- Department of Pediatrics, Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY.
| |
Collapse
|
5
|
Wang LH, Gao SZ, Bai XL, Chen ZL, Yang F. An Up-To-Date Overview of Dental Tissue Regeneration Using Dental Origin Mesenchymal Stem Cells: Challenges and Road Ahead. Front Bioeng Biotechnol 2022; 10:855396. [PMID: 35497335 PMCID: PMC9039056 DOI: 10.3389/fbioe.2022.855396] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/02/2022] [Indexed: 11/13/2022] Open
Abstract
Stem cells (SCs) research has experienced exponential growth in recent years. SC-based treatments can enhance the lives of people suffering from cardiac ischemia, Alzheimer’s disease, and regenerative drug conditions, like bone or loss of teeth. Numerous kinds of progenitor/SCs have been hypothesized to depend on their potential to regain and/or heal wounded tissue and partly recover organ function. Growing data suggest that SCs (SCs) are concentrated in functions and that particular tissues have more SCs. Dental tissues, in particular, are considered a significant cause of mesenchymal stem cells (MSCs) cells appropriate for tissue regeneration uses. Tissue regeneration and SCs biology have particular attention in dentistry because they may give a novel method for creating clinical material and/or tissue redevelopment. Dental pulp, dental papilla, periodontal ligament, and dental follicle contain mesenchymal SCs. Such SCs, which must be identified and cultivated in specific tissue culture environments, may be used in tissue engineering applications such as tooth tissue, nerve regeneration, and bone redevelopment. A new cause of SCs, induced pluripotent SCs, was successfully made from human somatic cells, enabling the generation of the patient and disease-specific SCs. The dental SC’s (DSCs) multipotency, rapid proliferation rate, and accessibility make it an ideal basis of MSC for tissue redevelopment. This article discusses current advances in tooth SC investigation and its possible application in tissue redevelopment.
Collapse
Affiliation(s)
- Lin-Hong Wang
- Center for Plastic & Reconstructive Surgery, Department of Stomatology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Si-Zhe Gao
- Department of Stomatology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiao-Lei Bai
- Institute of Basic Science and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Zheng-Lin Chen
- Hangzhou Junhe Regenerative Medicine Research Center, Hangzhou, China
| | - Fan Yang
- Center for Plastic & Reconstructive Surgery, Department of Stomatology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- *Correspondence: Fan Yang,
| |
Collapse
|
6
|
Segler A, Braun T, Fischer HS, Dukatz R, Weiss CR, Schwickert A, Jäger C, Bührer C, Henrich W. Feasibility of Umbilical Cord Blood Collection in Neonates at Risk of Brain Damage-A Step Toward Autologous Cell Therapy for a High-risk Population. Cell Transplant 2021; 30:963689721992065. [PMID: 33631961 PMCID: PMC7917411 DOI: 10.1177/0963689721992065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Evidence for umbilical cord blood (UCB) cell therapies as a potential intervention for neurological diseases is emerging. To date, most existing trials worked with allogenic cells, as the collection of autologous UCB from high-risk patients is challenging. In obstetric emergencies the collection cannot be planned. In preterm infants, late cord clamping and anatomic conditions may reduce the availability. The aim of the present study was to assess the feasibility of UCB collection in neonates at increased risk of brain damage. Infants from four high-risk groups were included: newborns with perinatal hypoxemia, gestational age (GA) ≤30 + 0 weeks and/or birthweight <1,500 g, intrauterine growth restriction (IUGR), or monochorionic twins with twin-to-twin transfusion syndrome (TTTS). Feasibility of collection, quantity and quality of obtained UCB [total nucleated cell count (TNC), volume, sterility, and cell viability], and neonatal outcome were assessed. UCB collection was successful in 141 of 177 enrolled patients (hypoxemia n = 10; GA ≤30 + 0 weeks n = 54; IUGR n = 71; TTTS n = 6). Twenty-six cases were missed. The amount of missed cases per month declined over the time. Volume of collected UCB ranged widely (median: 24.5 ml, range: 5.0–102 ml) and contained a median of 0.77 × 108 TNC (range: 0.01–13.0 × 108). TNC and UCB volume correlated significantly with GA. A total of 10.7% (19/177) of included neonates developed brain lesions. To conclude, collection of UCB in neonates at high risk of brain damage is feasible with a multidisciplinary approach and intensive training. High prevalence of brain damage makes UCB collection worthwhile. Collected autologous UCB from mature neonates harbors a sufficient cell count for potential therapy. However, quality and quantity of obtained UCB are critical for potential therapy in preterm infants. Therefore, for extremely preterm infants alternative cell sources such as UCB tissue should be investigated for autologous treatment options because of the low yield of UCB.
Collapse
Affiliation(s)
- Angela Segler
- Department of Obstetrics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Thorsten Braun
- Department of Obstetrics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of "Experimental Obstetrics" and Study group "Perinatal Programming", Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Hendrik Stefan Fischer
- Department of Neonatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ricarda Dukatz
- Department of Obstetrics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Claire-Rachel Weiss
- Department of Obstetrics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alexander Schwickert
- Department of Obstetrics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Carsten Jäger
- Department of Surgery, Klinikum rechts der Isar, Technische Universitaüt Muünchen, Munich, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Wolfgang Henrich
- Department of Obstetrics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
7
|
Anwar I, Ashfaq UA, Shokat Z. Therapeutic Potential of Umbilical Cord Stem Cells for Liver Regeneration. Curr Stem Cell Res Ther 2020; 15:219-232. [PMID: 32077830 DOI: 10.2174/1568026620666200220122536] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/16/2019] [Accepted: 08/08/2019] [Indexed: 01/18/2023]
Abstract
The liver is a vital organ for life and the only internal organ that is capable of natural regeneration. Although the liver has high regeneration capacity, excessive hepatocyte death can lead to liver failure. Various factors can lead to liver damage including drug abuse, some natural products, alcohol, hepatitis, and autoimmunity. Some models for studying liver injury are APAP-based model, Fas ligand (FasL), D-galactosamine/endotoxin (Gal/ET), Concanavalin A, and carbon tetrachloride-based models. The regeneration of the liver can be carried out using umbilical cord blood stem cells which have various advantages over other stem cell types used in liver transplantation. UCB-derived stem cells lack tumorigenicity, have karyotype stability and high immunomodulatory, low risk of graft versus host disease (GVHD), low risk of transmitting somatic mutations or viral infections, and low immunogenicity. They are readily available and their collection is safe and painless. This review focuses on recent development and modern trends in the use of umbilical cord stem cells for the regeneration of liver fibrosis.
Collapse
Affiliation(s)
- Ifrah Anwar
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Usman A Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Zeeshan Shokat
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| |
Collapse
|
8
|
Um S, Ha J, Choi SJ, Oh W, Jin HJ. Prospects for the therapeutic development of umbilical cord blood-derived mesenchymal stem cells. World J Stem Cells 2020; 12:1511-1528. [PMID: 33505598 PMCID: PMC7789129 DOI: 10.4252/wjsc.v12.i12.1511] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/23/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Umbilical cord blood (UCB) is a primitive and abundant source of mesenchymal stem cells (MSCs). UCB-derived MSCs have a broad and efficient therapeutic capacity to treat various diseases and disorders. Despite the high latent self-renewal and differentiation capacity of these cells, the safety, efficacy, and yield of MSCs expanded for ex vivo clinical applications remains a concern. However, immunomodulatory effects have emerged in various disease models, exhibiting specific mechanisms of action, such as cell migration and homing, angiogenesis, anti-apoptosis, proliferation, anti-cancer, anti-fibrosis, anti-inflammation and tissue regeneration. Herein, we review the current literature pertaining to the UCB-derived MSC application as potential treatment strategies, and discuss the concerns regarding the safety and mass production issues in future applications.
Collapse
Affiliation(s)
- Soyoun Um
- Research Team for Immune Cell Therapy, Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| | - Jueun Ha
- Research Team for Osteoarthritis, Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| | - Wonil Oh
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| | - Hye Jin Jin
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| |
Collapse
|
9
|
Haque N, Khan IM, Abu Kasim NH. Survival and immunomodulation of stem cells from human extracted deciduous teeth expanded in pooled human and foetal bovine sera. Cytokine 2019; 120:144-154. [PMID: 31071675 DOI: 10.1016/j.cyto.2019.04.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/20/2019] [Accepted: 04/27/2019] [Indexed: 12/17/2022]
Abstract
The immunomodulatory properties of mesenchymal stem cells (MSCs) from autologous and allogeneic sources are useful in stimulating tissue regeneration and repair. To obtain a high number of MSCs for transplantation requires extensive in vitro expansion with culture media supplements that can cause xeno-contamination of cells potentially compromising function and clinical outcomes. In this study stem cells from human extracted deciduous teeth (SHED) were cultured in Knockout™ DMEM supplemented with either pooled human serum (pHS) or foetal bovine serum (FBS) to compare their suitability in maintaining immunomodulatory properties of cells during in vitro expansion. No significant difference in cell survival of SHED grown in pHS (pHS-SHED) or FBS (FBS-SHED) was observed when co-cultured with complement, monocytes or lymphocytes. However, significant changes in the expression of sixteen paracrine factors involved in immunomodulation were observed in the supernatants of FBS-SHED co-cultures with monocytes or lymphocytes compared to that in pHS-SHEDs after both 24 and 120 h of incubation. Further analysis of changing protein levels of paracrine factors in co-cultures using biological pathway analysis software predicted upregulation of functions associated with immunogenicity in FBS-SHED and lymphocyte co-cultures compared to pHS-SHED co-cultures. Pathway analysis also predicted significant stimulation of HMGB1 and TREM1 signalling pathways in FBS-SHED co-cultures indicating activation of immune cells and inflammation. Though FBS supplementation does not impact survival of SHED, our combinatorial biological pathway analysis supports the idea that in vitro expansion of SHEDs in pHS provides optimal conditions to minimise xeno-contamination and inflammation and maintain their immunomodulatory properties.
Collapse
Affiliation(s)
- Nazmul Haque
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Selangor, Malaysia; Regenerative Dentistry Research Group, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Ilyas M Khan
- Centre for NanoHealth, Swansea University Medical School, Swansea, UK
| | - Noor Hayaty Abu Kasim
- Regenerative Dentistry Research Group, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia; Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
10
|
Ciubotariu R, Scaradavou A, Ciubotariu I, Tarnawski M, Lloyd S, Albano M, Dobrila L, Rubinstein P, Grunebaum A. Impact of delayed umbilical cord clamping on public cord blood donations: can we help future patients and benefit infant donors? Transfusion 2018; 58:1427-1433. [PMID: 29574750 DOI: 10.1111/trf.14574] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/11/2017] [Accepted: 01/22/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Cord blood (CB) is a widely accepted stem cell source and its clinical utilization depends, to a great extent, on its cell content. Birth-to-clamping (BTC) time of umbilical cord determines placental transfusion to the newborn, and the remaining blood that can be collected and banked. The 2017 Committee Opinion of the American College of Obstetrics and Gynecologists (ACOG) recommends a delay of "at least 30-60 seconds" before clamping the cord for all newborns to ensure adequate iron stores. The impact of delayed cord clamping (DCC) on public CB banking can be substantial. STUDY DESIGN AND METHODS Cord blood units (CBUs) collected from 1210 mothers at one hospital were evaluated for total nucleated cells (TNCs) and weight/volume based on time to clamping. Bank staff recorded BTC time in seconds as reported by obstetricians; collections were performed ex utero. Immediate clamping was defined as BTC of less than 30 seconds, whereas DCC was defined as BTC of 30 seconds or more. RESULTS Cord clamping was immediate in 903 (75%) and delayed in 307 (25%) deliveries. Successful recovery (% clinical CBUs) decreased 10-fold with DCC of more than 60 seconds (22% vs. 2.4%, p < 0.001). CBUs collected after DCC of more than 60 seconds had significantly lower TNC counts than those after DCC of less than 60 seconds (p < 0.0001). Furthermore, 38% to 46% of CBUs after DCC of more than 60 seconds had volume of less than 40 mL. CONCLUSION Our study indicates that DCC of 30 to 60 seconds has a small negative impact on collection of high-TNC-count CBUs. However, increasing BTC to more than 60 seconds decreases significantly both TNC content and volume, reducing drastically the chances of obtaining clinically useful CBUs.
Collapse
Affiliation(s)
- Rodica Ciubotariu
- National Cord Blood Program, New York Blood Center, New York, New York
| | | | | | - Michal Tarnawski
- National Cord Blood Program, New York Blood Center, New York, New York
| | - Sara Lloyd
- National Cord Blood Program, New York Blood Center, New York, New York
| | - Maria Albano
- National Cord Blood Program, New York Blood Center, New York, New York
| | - Ludy Dobrila
- National Cord Blood Program, New York Blood Center, New York, New York
| | - Pablo Rubinstein
- National Cord Blood Program, New York Blood Center, New York, New York
| | - Amos Grunebaum
- New York Presbyterian Hospital/Weill Cornell Medical Center, New York, New York
| |
Collapse
|
11
|
Roura S, Gálvez-Montón C, Mirabel C, Vives J, Bayes-Genis A. Mesenchymal stem cells for cardiac repair: are the actors ready for the clinical scenario? Stem Cell Res Ther 2017; 8:238. [PMID: 29078809 PMCID: PMC5658929 DOI: 10.1186/s13287-017-0695-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
For years, sufficient progress has been made in treating heart failure following myocardial infarction; however, the social and economic burdens and the costs to world health systems remain high. Moreover, treatment advances have not resolved the underlying problem of functional heart tissue loss. In this field of research, for years we have actively explored innovative biotherapies for cardiac repair. Here, we present a general, critical overview of our experience in using mesenchymal stem cells, derived from cardiac adipose tissue and umbilical cord blood, in a variety of cell therapy and tissue engineering approaches. We also include the latest advances and future challenges, including good manufacturing practice and regulatory issues. Finally, we evaluate whether recent approaches hold potential for reliable translation to clinical trials.
Collapse
Affiliation(s)
- Santiago Roura
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Badalona, Spain. .,Center of Regenerative Medicine in Barcelona, Barcelona, Spain. .,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain. .,ICREC (Heart Failure and Cardiac Regeneration) Research Programme, Health Sciences Research Institute Germans Trias i Pujol (IGTP), Carretera de Can Ruti, Camí de les Escoles s/n, 08916, Badalona, Barcelona, Spain.
| | - Carolina Gálvez-Montón
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Clémentine Mirabel
- Servei de Teràpia Cel∙lular, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005, Barcelona, Spain.,Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron 129-139, 08035, Barcelona, Spain
| | - Joaquim Vives
- Servei de Teràpia Cel∙lular, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005, Barcelona, Spain.,Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron 129-139, 08035, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Antoni Bayes-Genis
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Badalona, Spain. .,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain. .,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain. .,Cardiology Service, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain. .,Heart Institute, Hospital Universitari Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916, Badalona, Barcelona, Spain.
| |
Collapse
|
12
|
Sordi V, Pellegrini S, Krampera M, Marchetti P, Pessina A, Ciardelli G, Fadini G, Pintus C, Pantè G, Piemonti L. Stem cells to restore insulin production and cure diabetes. Nutr Metab Cardiovasc Dis 2017; 27:583-600. [PMID: 28545927 DOI: 10.1016/j.numecd.2017.02.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 01/24/2017] [Accepted: 02/11/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND The advancement of knowledge in the field of regenerative medicine is increasing the therapeutic expectations of patients and clinicians on cell therapy approaches. Within these, stem cell therapies are often evoked as a possible therapeutic option for diabetes, already ongoing or possible in the near future. AIM The purpose of this document is to make a point of the situation on existing knowledge and therapies with stem cells to treat patients with diabetes by focusing on some of the aspects that most frequently raise curiosity and discussion in clinical practice and in the interaction with the patient. In fact, at present there are no clinically approved treatments based on the use of stem cells for the treatment of diabetes, but several therapeutic approaches have already been evaluated or are being evaluated in clinical trials. DATA SYNTHESIS It is possible to identify three large potential application fields: 1) the reconstruction of the β cell mass; 2) the immunomodulation in type 1 diabetes (T1D); 3) the treatment of complications. In this study we will limit the discussion to approaches that have the potential for clinical translation, deliberately omitting aspects of basic biology and preclinical data. Also, we intentionally omit the treatment of the complications that will be the subject of a future document. Finally, an overview of the Italian situation regarding the storage of cord blood cells for the therapy of diabetes will be given.
Collapse
Affiliation(s)
- V Sordi
- Diabetes Research Institute (DRI) - IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - S Pellegrini
- Diabetes Research Institute (DRI) - IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - M Krampera
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Italy
| | - P Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - A Pessina
- CRC-StaMeTec (Mesenchymal Stem Cells for Cell Therapy), Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - G Ciardelli
- DIMEAS - Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - G Fadini
- Medicine Department (DIMED), University of Padua, Italy
| | - C Pintus
- Italian National Transplant Center (CNT), Italy
| | - G Pantè
- Italian Medicines Agency (AIFA), Italy
| | - L Piemonti
- Diabetes Research Institute (DRI) - IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
13
|
Rashidghamat E, McGrath JA. Novel and emerging therapies in the treatment of recessive dystrophic epidermolysis bullosa. Intractable Rare Dis Res 2017; 6:6-20. [PMID: 28357176 PMCID: PMC5359356 DOI: 10.5582/irdr.2017.01005] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Epidermolysis bullosa (EB) is a clinically and genetically heterogeneous group of inherited blistering diseases that affects ∼ 500,000 people worldwide. Clinically, individuals with EB have fragile skin and are susceptible to blistering following minimal trauma, with mucous membrane and other organ involvement in some subtypes. Within the spectrum of EB, ∼ 5% of affected individuals have the clinically more severe recessive dystrophic (RDEB) variant with a prevalence of 8 per one million of the population. RDEB is caused by loss-of-function mutations in the type VII collagen gene, COL7A1, which leads to reduced or absent type VII collagen (C7) and a paucity of structurally effective anchoring fibrils at the dermal-epidermal junction (DEJ). Currently, there is no cure for RDEB, although considerable progress has been made in testing novel treatments including gene therapy (lentiviral and gamma retroviral vectors for COL7A1 supplementation in keratinocytes and fibroblasts), as well as cell therapy (use of allogeneic fibroblasts, mesenchymal stromal cells (MSCs), and bone marrow transplantation (BMT)). Here, we review current treatment modalities available as well as novel and emerging therapies in the treatment of RDEB. Clinical trials of new translational therapies in RDEB offer hope for improved clinical management of patients as well as generating broader lessons for regenerative medicine that could be applicable to other inherited or acquired abnormalities of wound healing or scarring.
Collapse
Affiliation(s)
- Ellie Rashidghamat
- St. John's Institute of Dermatology, King's College London, London, United Kingdom
| | - John A. McGrath
- St. John's Institute of Dermatology, King's College London, London, United Kingdom
- Address correspondence to: Dr. John A. McGrath, Dermatology Research Laboratories, Floor 9 Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom. E-mail:
| |
Collapse
|
14
|
Genetically re-engineered K562 cells significantly expand and functionally activate cord blood natural killer cells: Potential for adoptive cellular immunotherapy. Exp Hematol 2016; 46:38-47. [PMID: 27765614 DOI: 10.1016/j.exphem.2016.10.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 09/19/2016] [Accepted: 10/07/2016] [Indexed: 01/11/2023]
Abstract
Natural killer (NK) cells play a significant role in reducing relapse in patients with hematological malignancies after allogeneic stem cell transplantation, but NK cell number and naturally occurring inhibitory signals limit their capability. Interleukin-15 (IL-15) and 4-1BBL are important modulators of NK expansion and functional activation. To overcome these limitations, cord blood mononuclear cells (CB MNCs) were ex vivo expanded for 7 days with genetically modified K562-mbIL15-41BBL (MODK562) or wild-type K562 (WTK562). NK cell expansion; expression of lysosome-associated membrane protein-1 (LAMP-1), granzyme B, and perforin; and in vitro and in vivo cytotoxicity against B-cell non-Hodgkin lymphoma (B-NHL) were evaluated. In vivo tumor growth in B-NHL-xenografted nonobese diabetic severe combined immune deficient (NOD-scid) gamma (NSG) mice was monitored by tumor volume, cell number, and survival. CB MNCs cultured with MODK562 compared with WTK562 demonstrated significantly increased NK expansion (thirty-fivefold, p < 0.05); LAMP-1 (p < 0.05), granzyme B, and perforin expression (p < 0.001); and in vitro cytotoxicity against B-NHL (p < 0.01). Xenografted mice treated with MODK562 CB experienced significantly decreased B-NHL tumor volume (p = 0.0086) and B-NHL cell numbers (p < 0.01) at 5 weeks and significantly increased survival (p < 0.001) at 10 weeks compared with WTK562. In summary, MODK562 significantly enhanced CB NK expansion and cytotoxicity, enhanced survival in a human Burkitt's lymphoma xenograft NSG model, and could be used in the future as adoptive cellular immunotherapy after umbilical CB transplantation. Future directions include expanding anti-CD20 chimeric receptor-modified CB NK cells to enhance B-NHL targeting in vitro and in vivo.
Collapse
|
15
|
Stem Cell Banking and Its Impact on Cardiac Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 951:163-178. [PMID: 27837563 DOI: 10.1007/978-3-319-45457-3_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Cardiovascular diseases, including heart failure, are the most frequent cause of death annually, even higher than any other pathologies. Specifically, patients who suffer from myocardial infarction may encounter adverse remodeling processes of the heart that can ultimately lead to heart failure. Prognosis of patients affected by heart failure is very poor with 5-year mortality close to 50 %. Despite the impressive progress in the clinical treatment of heart failure in recent years, heart transplantation is still required to avoid death as the result of the inexorable decline in cardiac function. Unfortunately, the availability of donor human hearts for transplantation largely fails to cover the number of potential recipient requests. From this urgent unmet clinical need the interest in stem cell applications for heart regeneration made its start, and has rapidly grown in the last decades. Indeed, the discovery and application of stem and progenitor cells as therapeutic agents has raised substantial interest with the objective of reversing these processes, and ultimately inducing cardiac regeneration. In this scenario, the role of biobanking may play a remarkable role to provide cells at the right time according to the patient's clinical needs, mostly for autologous use in the acute setting of myocardial infarction, largely reducing the time needed for cell preparation and expansion before administration.
Collapse
|
16
|
Zanolini D, Merlin S, Feola M, Ranaldo G, Amoruso A, Gaidano G, Zaffaroni M, Ferrero A, Brunelleschi S, Valente G, Gupta S, Prat M, Follenzi A. Extrahepatic sources of factor VIII potentially contribute to the coagulation cascade correcting the bleeding phenotype of mice with hemophilia A. Haematologica 2015; 100:881-92. [PMID: 25911555 DOI: 10.3324/haematol.2014.123117] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 04/22/2015] [Indexed: 12/14/2022] Open
Abstract
A large fraction of factor VIII in blood originates from liver sinusoidal endothelial cells although extrahepatic sources also contribute to plasma factor VIII levels. Identification of cell-types other than endothelial cells with the capacity to synthesize and release factor VIII will be helpful for therapeutic approaches in hemophilia A. Recent cell therapy and bone marrow transplantation studies indicated that Küpffer cells, monocytes and mesenchymal stromal cells could synthesize factor VIII in sufficient amount to ameliorate the bleeding phenotype in hemophilic mice. To further establish the role of blood cells in expressing factor VIII, we studied various types of mouse and human hematopoietic cells. We identified factor VIII in cells isolated from peripheral and cord blood, as well as bone marrow. Co-staining for cell type-specific markers verified that factor VIII was expressed in monocytes, macrophages and megakaryocytes. We additionally verified that factor VIII was expressed in liver sinusoidal endothelial cells and endothelial cells elsewhere, e.g., in the spleen, lungs and kidneys. Factor VIII was well expressed in sinusoidal endothelial cells and Küpffer cells isolated from human liver, whereas by comparison isolated human hepatocytes expressed factor VIII at very low levels. After transplantation of CD34(+) human cord blood cells into NOD/SCIDγNull-hemophilia A mice, fluorescence activated cell sorting of peripheral blood showed >40% donor cells engrafted in the majority of mice. In these animals, plasma factor VIII activity 12 weeks after cell transplantation was up to 5% and nine of 12 mice survived after a tail clip-assay. In conclusion, hematopoietic cells, in addition to endothelial cells, express and secrete factor VIII: this information should offer further opportunities for understanding mechanisms of factor VIII synthesis and replenishment.
Collapse
Affiliation(s)
- Diego Zanolini
- Dept. of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Simone Merlin
- Dept. of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Maria Feola
- Dept. of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Gabriella Ranaldo
- Dept. of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Angela Amoruso
- Dept. of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Gianluca Gaidano
- Dept. of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Mauro Zaffaroni
- Azienda Ospedaliera Universitaria Maggiore della Carità, SCDU Pediatria, Novara, Italy
| | - Alessandro Ferrero
- Azienda Ospedaliera Mauriziano, SC Chirurgia Generale ed Oncologica, Torino, Italy
| | - Sandra Brunelleschi
- Dept. of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Guido Valente
- Dept. of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Sanjeev Gupta
- Dept. of Medicine and Pathology, Marion Bessin Liver Research Center, Diabetes Center, Cancer Center, and Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maria Prat
- Dept. of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Antonia Follenzi
- Dept. of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| |
Collapse
|
17
|
Insights and hopes in umbilical cord blood stem cell transplantations. J Biomed Biotechnol 2012; 2012:572821. [PMID: 23258957 PMCID: PMC3509718 DOI: 10.1155/2012/572821] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2012] [Accepted: 10/08/2012] [Indexed: 11/17/2022] Open
Abstract
Over 20.000 umblical cord blood transplantations (UCBT) have been carried out around the world. Indeed, UCBT represents an attractive source of donor hematopoietic stem cells (HSCs) and, offer interesting features (e.g., lower graft-versus-host disease) compared to bone marrow transplantation (BMT). Thereby, UCBT often represents the unique curative option against several blood diseases. Recent advances in the field of UCBT, consisted to develop strategies to expand umbilical stem cells and shorter the timing of their engraftment, subsequently enhancing their availability for enhanced efficacy of transplantation into indicated patients with malignant diseases (e.g., leukemia) or non-malignant diseases (e.g., thalassemia major). Several studies showed that the expansion and homing of UCBSCs depends on specific biological factors and cell types (e.g., cytokines, neuropeptides, co-culture with stromal cells). In this review, we extensively present the advantages and disadvantages of current hematopoietic stem cell transplantations (HSCTs), compared to UBCT. We further describe the importance of cord blood content and obstetric factors on cord blood selection, and report the recent approaches that can be undertook to improve cord blood stem cell expansion as well as engraftment. Eventually, we provide two majors examples underlining the importance of UCBT as a potential cure for blood diseases.
Collapse
|
18
|
Rescuing the neonatal brain from hypoxic injury with autologous cord blood. Bone Marrow Transplant 2012; 48:890-900. [PMID: 22964590 DOI: 10.1038/bmt.2012.169] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 08/02/2012] [Accepted: 08/03/2012] [Indexed: 12/28/2022]
Abstract
Brain injury resulting from perinatal hypoxic-ischemic encephalopathy (HIE) is a major cause of acute mortality in infants and chronic neurologic disability in surviving children. Recent multicenter clinical trials demonstrated the effectiveness of hypothermia initiated within the first 6 postnatal hours to reduce the risk of death or major neurological disabilities among neonates with HIE. However, in these trials, approximately 40% of cooled infants died or survived with significant impairments. Therefore, adjunct therapies are required to improve the outcome in neonates with HIE. Cord blood (CB) is a rich source of stem cells. Administration of human CB cells in animal models of HIE has generally resulted in improved outcomes and multiple mechanisms have been suggested including anti-inflammation, release of neurotrophic factors and stimulation of endogenous neurogenesis. Investigators at Duke are conducting studies of autologous CB infusion in neonates with HIE and in children with cerebral palsy. These pilot studies indicate no added risk from the regimens used, but results of ongoing placebo-controlled trials are needed to assess efficacy. Meanwhile, further investigations are warranted to determine the best strategies, that is, timing, dosing, route of delivery, choice of stem cells and ex vivo modulations, to attain long-term benefits of CB stem cell therapy.
Collapse
|
19
|
Ferreira MSV, Schneider RK, Wagner W, Jahnen-Dechent W, Labude N, Bovi M, Piroth D, Knüchel R, Hieronymus T, Müller AM, Zenke M, Neuss S. Two-dimensional polymer-based cultures expand cord blood-derived hematopoietic stem cells and support engraftment of NSG mice. Tissue Eng Part C Methods 2012; 19:25-38. [PMID: 22712684 DOI: 10.1089/ten.tec.2011.0706] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Currently, ex vivo expansion of hematopoietic stem cells (HSC) is still insufficient. Traditional approaches for HSC expansion include the use of stromal cultures, growth factors, and/or bioreactors. Biomaterial-based strategies provide new perspectives. We focus on identifying promising two-dimensional (2D) polymer candidates for HSC expansion. After a 7-day culture period with cytokine supplementation, 2D fibrin, poly(D,L-lactic-co-glycolic acid; Resomer® RG503), and Poly(ɛ-caprolactone; PCL) substrates supported expansion of cord blood (CB)-derived CD34⁺ cells ex vivo. Fibrin cultures achieved the highest proliferation rates (>8700-fold increase of total nuclear cells, p<0.001), high total colony-forming units (3.6-fold increase, p<0.001), and highest engraftment in NSG mice (7.69-fold more donor cells compared with tissue culture polysterene, p<0.001). In addition, the presence of multiple human hematopoietic lineages such as myeloid (CD13⁺), erythroid (GypC⁺), and lymphoid (CD20⁺/CD56⁺) in murine transplant recipients confirmed the multilineage engraftment potential of fibrin-based cultures. Filopodia development in fibrin-expanded cells was a further indicator for superior cell adhesion capacities. We propose application of fibrin, Resomer® RG503, and PCL for future strategies of CB-CD34⁺ cell expansion. Suitable polymers for HSC expansion might also be appropriate for future drug discovery applications or for studies aimed to develop hematological therapies.
Collapse
|
20
|
Jin HL, Kim JS, Kim YJ, Kim SJ, Broxmeyer HE, Kim KS. Dynamic expression of specific miRNAs during erythroid differentiation of human embryonic stem cells. Mol Cells 2012; 34:177-83. [PMID: 22767248 PMCID: PMC3887816 DOI: 10.1007/s10059-012-0090-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 04/16/2012] [Accepted: 05/16/2012] [Indexed: 11/25/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression at post-transcriptional levels through mRNA degradation or translation inhibition. Little is known regarding miRNA participation in regulating hematopoietic, or more specifically erythroid differentiation. This study was aimed at identifying erythroid lineage-specific miRNAs expressed during in vitro erythropoiesis using human embryonic stem cells (hESCs) and human umbilical cord blood (CB) CD34+ cells. CD34+ hematopoietic cells were produced from hESCs in vitro and subsequently induced to differentiate into erythroid cells by culture in sequence on OP9 feeder cells and then with mesenchymal stromal cells (MSC) in the presence of cytokines. Expression profiles of erythroid lineage-specific miRNAs were analyzed by quantitative PCR during in vitro differentiation. Expression levels of miR-142-3p, miR-142-5p, miR-146a and miR-451 were dynamically changed during differentiation of hESCs to CD34+ hematopoietic cells, and in subsequent differentiation of the CD34+ cells into the erythroid lineage. This suggests that these four miRNAs might be involved in regulating erythropoiesis.
Collapse
Affiliation(s)
- Hong Lian Jin
- Graduate School of Biomedical Science and Engineering, College of Medicine, Department of Anatomy and Cell Biology, Hanyang University, Seoul 133-791,
Korea
| | - Jong Soo Kim
- Graduate School of Biomedical Science and Engineering, College of Medicine, Department of Anatomy and Cell Biology, Hanyang University, Seoul 133-791,
Korea
| | - Young June Kim
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis 46202,
USA
| | - Su Jin Kim
- Graduate School of Biomedical Science and Engineering, College of Medicine, Department of Anatomy and Cell Biology, Hanyang University, Seoul 133-791,
Korea
| | - Hal E. Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis 46202,
USA
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, College of Medicine, Department of Anatomy and Cell Biology, Hanyang University, Seoul 133-791,
Korea
| |
Collapse
|
21
|
Roura S, Pujal JM, Bayes-Genis A. Umbilical cord blood for cardiovascular cell therapy: from promise to fact. Ann N Y Acad Sci 2012; 1254:66-70. [PMID: 22548571 DOI: 10.1111/j.1749-6632.2012.06515.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Endothelial recovery and cell replacement are therapeutic challenges for cardiovascular medicine. Initially employed in the treatment of blood malignancies due to its high concentration of hematological precursors, umbilical cord blood (UCB) is now a non-controversial and accepted source of both hematopoietic and non-hematopoietic progenitors for a variety of emerging cell therapies in clinical trials. Here, we review the current therapeutic potential of UCB, focusing in recent evidence demonstrating the ability of UCB-derived mesenchymal stem cells to differentiate into the endothelial lineage and to develop new vasculature in vivo.
Collapse
Affiliation(s)
- Santiago Roura
- ICREC Research Group, Health Sciences Research Institute Germans Trias i Pujol (IGTP), Barcelona, Spain
| | | | | |
Collapse
|
22
|
Gluckman E, Ruggeri A, Rocha V, Baudoux E, Boo M, Kurtzberg J, Welte K, Navarrete C, van Walraven SM. Family-directed umbilical cord blood banking. Haematologica 2011; 96:1700-7. [PMID: 21750089 PMCID: PMC3208689 DOI: 10.3324/haematol.2011.047050] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 06/16/2011] [Accepted: 06/30/2011] [Indexed: 01/21/2023] Open
Abstract
Umbilical cord blood transplantation from HLA-identical siblings provides good results in children. These results support targeted efforts to bank family cord blood units that can be used for a sibling diagnosed with a disease which can be cured by allogeneic hematopoietic stem cell transplantation or for research that investigates the use of allogeneic or autologous cord blood cells. Over 500 patients transplanted with related cord blood units have been reported to the Eurocord registry with a 4-year overall survival of 91% for patients with non-malignant diseases and 56% for patients with malignant diseases. Main hematologic indications in children are leukemia, hemoglobinopathies or inherited hematologic, immunological or metabolic disorders. However, family-directed cord blood banking is not widely promoted; many cord blood units used in sibling transplantation have been obtained from private banks that do not meet the necessary criteria required to store these units. Marketing by private banks who predominantly store autologous cord blood units has created public confusion. There are very few current validated indications for autologous storage but some new indications might appear in the future. Little effort is devoted to provide unbiased information and to educate the public as to the distinction between the different types of banking, economic models and standards involved in such programs. In order to provide a better service for families in need, directed-family cord blood banking activities should be encouraged and closely monitored with common standards, and better information on current and future indications should be made available.
Collapse
Affiliation(s)
- Eliane Gluckman
- Eurocord, Hospital Saint Louis, University Paris VII, Paris, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Geyer MB, Jacobson JS, Freedman J, George D, Moore V, van de Ven C, Satwani P, Bhatia M, Garvin JH, Bradley MB, Harrison L, Morris E, Della-Latta P, Schwartz J, Baxter-Lowe LA, Cairo MS. A comparison of immune reconstitution and graft-versus-host disease following myeloablative conditioning versus reduced toxicity conditioning and umbilical cord blood transplantation in paediatric recipients. Br J Haematol 2011; 155:218-34. [PMID: 21848882 DOI: 10.1111/j.1365-2141.2011.08822.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Immune reconstitution appears to be delayed following myeloablative conditioning (MAC) and umbilical cord blood transplantation (UCBT) in paediatric recipients. Although reduced toxicity conditioning (RTC) versus MAC prior to allogeneic stem cell transplantation is associated with decreased transplant-related mortality, the effects of RTC versus MAC prior to UCBT on immune reconstitution and risk of graft-versus-host disease (GVHD) are unknown. In 88 consecutive paediatric recipients of UCBT, we assessed immune cell recovery and immunoglobulin reconstitution at days +100, 180 and 365 and analysed risk factors associated with acute and chronic GVHD. Immune cell subset recovery, immunoglobulin reconstitution, and the incidence of opportunistic infections did not differ significantly between MAC versus RTC groups. In a Cox model, MAC versus RTC recipients had significantly higher risk of grade II-IV acute GVHD [Hazard Ratio (HR) 6·1, P = 0·002] as did recipients of 4/6 vs. 5-6/6 HLA-matched UCBT (HR 3·1, P = 0·03), who also had significantly increased risk of chronic GVHD (HR 18·5, P = 0·04). In multivariate analyses, MAC versus RTC was furthermore associated with significantly increased transplant-related (Odds Ratio 26·8, P = 0·008) and overall mortality (HR = 4·1, P = 0·0001). The use of adoptive cellular immunotherapy to accelerate immune reconstitution and prevent and treat opportunistic infections and malignant relapse following UCBT warrants further investigation.
Collapse
Affiliation(s)
- Mark B Geyer
- Department of Pediatrics, NewYork-Presbyterian Morgan Stanley Children's Hospital, Columbia University, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Estrela C, Alencar AHGD, Kitten GT, Vencio EF, Gava E. Mesenchymal stem cells in the dental tissues: perspectives for tissue regeneration. Braz Dent J 2011; 22:91-8. [DOI: 10.1590/s0103-64402011000200001] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 02/28/2011] [Indexed: 01/02/2023] Open
Abstract
In recent years, stem cell research has grown exponentially owing to the recognition that stem cell-based therapies have the potential to improve the life of patients with conditions that range from Alzheimer’s disease to cardiac ischemia and regenerative medicine, like bone or tooth loss. Based on their ability to rescue and/or repair injured tissue and partially restore organ function, multiple types of stem/progenitor cells have been speculated. Growing evidence demonstrates that stem cells are primarily found in niches and that certain tissues contain more stem cells than others. Among these tissues, the dental tissues are considered a rich source of mesenchymal stem cells that are suitable for tissue engineering applications. It is known that these stem cells have the potential to differentiate into several cell types, including odontoblasts, neural progenitors, osteoblasts, chondrocytes, and adipocytes. In dentistry, stem cell biology and tissue engineering are of great interest since may provide an innovative for generation of clinical material and/or tissue regeneration. Mesenchymal stem cells were demonstrated in dental tissues, including dental pulp, periodontal ligament, dental papilla, and dental follicle. These stem cells can be isolated and grown under defined tissue culture conditions, and are potential cells for use in tissue engineering, including, dental tissue, nerves and bone regeneration. More recently, another source of stem cell has been successfully generated from human somatic cells into a pluripotent stage, the induced pluripotent stem cells (iPS cells), allowing creation of patient- and disease-specific stem cells. Collectively, the multipotency, high proliferation rates, and accessibility make the dental stem cell an attractive source of mesenchymal stem cells for tissue regeneration. This review describes new findings in the field of dental stem cell research and on their potential use in the tissue regeneration.
Collapse
Affiliation(s)
| | | | | | | | - Elisandra Gava
- Federal University of Goiás, Brazil; Federal University of Minas Gerais, Brazil
| |
Collapse
|
25
|
|