1
|
Aouabdi S, Aboalola D, Zakari S, Alwafi S, Nedjadi T, Alsiary R. Protective potential of mesenchymal stem cells against COVID-19 during pregnancy. Future Sci OA 2024; 10:FSO924. [PMID: 38836262 PMCID: PMC11149780 DOI: 10.2144/fsoa-2023-0179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/17/2023] [Indexed: 06/06/2024] Open
Abstract
SARS-CoV-2 causes COVID-19. COVID-19 has led to severe clinical illnesses and an unprecedented death toll. The virus induces immune inflammatory responses specifically cytokine storm in lungs. Several published reports indicated that pregnant females are less likely to develop severe symptoms compared with non-pregnant. Putative protective role of maternal blood circulating fetal mesenchymal stem cells (MSCs) has emerged and have been put forward as an explanation to alleviated symptoms. MSCs with immune-modulatory, anti-inflammatory and anti-viral roles, hold great potential for the treatment of COVID-19. MSCs could be an alternative to treat infections resulting from the SARS-CoV-2 and potential future outbreaks. This review focuses on the MSCs putative protective roles against COVID-19 in pregnant females.
Collapse
Affiliation(s)
- Sihem Aouabdi
- King Abdullah International Medical Research Center, Jeddah, 21423, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, 21423, Saudi Arabia
| | - Doaa Aboalola
- King Abdullah International Medical Research Center, Jeddah, 21423, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, 21423, Saudi Arabia
| | - Samer Zakari
- King Abdullah International Medical Research Center, Jeddah, 21423, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, 21423, Saudi Arabia
| | - Suliman Alwafi
- King Abdullah International Medical Research Center, Jeddah, 21423, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, 21423, Saudi Arabia
| | - Taoufik Nedjadi
- King Abdullah International Medical Research Center, Jeddah, 21423, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, 21423, Saudi Arabia
| | - Rawiah Alsiary
- King Abdullah International Medical Research Center, Jeddah, 21423, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, 21423, Saudi Arabia
| |
Collapse
|
2
|
Mikkelsen E, Huppertz B, Singh R, Ravn K, Hatt L, Kruhøffer M, Urrabaz-Garza R, Uldbjerg N, Menon R, Steiniche T. mRNA and Protein Expression in Human Fetal Membrane Cells: Potential Biomarkers for Preterm Prelabor Rupture of the Fetal Membranes? Int J Mol Sci 2023; 24:15826. [PMID: 37958809 PMCID: PMC10650701 DOI: 10.3390/ijms242115826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/27/2023] [Accepted: 10/28/2023] [Indexed: 11/15/2023] Open
Abstract
Clinically, unique markers in fetal membrane cells may contribute to the search for biomarkers for preterm prelabor rupture of the fetal membranes (pPROM) in maternal blood. pPROM is associated with overwhelming inflammation and premature cellular senescence causing "biological microfractures" of the fetal membranes. We hypothesize that these pathological processes are associated with the shedding of fetal membrane cells into the maternal circulation. The aim of this study was to identify markers expressed exclusively in fetal membrane cells to facilitate their isolation, characterization, and determination of biomarker potential in maternal blood. We have (1), by their transcriptomic profile, identified markers that are upregulated in amnion and chorion tissue compared to maternal white blood cells, and (2), by immunohistochemistry, confirmed the localization of the differentially expressed proteins in fetal membranes, placenta, and the placental bed of the uterus. RNA sequencing revealed 31 transcripts in the amnion and 42 transcripts in the chorion that were upregulated. Among these, 22 proteins were evaluated by immunohistochemistry. All but two transcripts were expressed both on mRNA and protein level in at least one fetal membrane cell type. Among these remaining 20 proteins, 9 proteins were not significantly expressed in the villous and extravillous trophoblasts of the placenta.
Collapse
Affiliation(s)
- Emmeli Mikkelsen
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Blvd. 11, 8200 Aarhus, Denmark; (E.M.); (N.U.)
- Department of Obstetrics and Gynaecology, Aarhus University Hospital, Palle Juul-Jensens Blvd. 99, 8200 Aarhus, Denmark
| | - Berthold Huppertz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria;
| | - Ripudaman Singh
- ARCEDI Biotech Aps, Tabletvej 1, 7100 Vejle, Denmark; (R.S.); (K.R.); (L.H.)
| | - Katarina Ravn
- ARCEDI Biotech Aps, Tabletvej 1, 7100 Vejle, Denmark; (R.S.); (K.R.); (L.H.)
| | - Lotte Hatt
- ARCEDI Biotech Aps, Tabletvej 1, 7100 Vejle, Denmark; (R.S.); (K.R.); (L.H.)
| | | | - Rheanna Urrabaz-Garza
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA (R.M.)
| | - Niels Uldbjerg
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Blvd. 11, 8200 Aarhus, Denmark; (E.M.); (N.U.)
- Department of Obstetrics and Gynaecology, Aarhus University Hospital, Palle Juul-Jensens Blvd. 99, 8200 Aarhus, Denmark
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA (R.M.)
| | - Torben Steiniche
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Blvd. 11, 8200 Aarhus, Denmark; (E.M.); (N.U.)
- Department of Histopathology, Aarhus University Hospital, Palle Juul-Jensens Blvd. 99, 8200 Aarhus, Denmark
| |
Collapse
|
3
|
Alkobtawi M, Sbeih M, Souaid K, Ngô QT, Nassar D, Arbes H, Guillet H, Habibi A, Bartolucci P, Castela M, Aractingi S, Oulès B. Contribution of fetal microchimeric cells to maternal wound healing in sickle cell ulcers. Haematologica 2023; 108:1920-1933. [PMID: 36373248 PMCID: PMC10316260 DOI: 10.3324/haematol.2022.281140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 10/31/2022] [Indexed: 08/18/2024] Open
Abstract
Leg ulcers are a major complication of sickle cell disease (SCD). They are particularly challenging to treat and innovative therapies are needed. We previously showed that the healing of SCD ulcers is delayed because of decreased angiogenesis. During pregnancy, fetal microchimeric cells (FMC) transferred to the mother are recruited to maternal wounds and improve angiogenesis. After delivery, FMC persist in maternal bone marrow for decades. Here, we investigated whether fetal cells could also improve SCD ulcers in the post-partum setting. We found that skin healing was similarly improved in post-partum mice and in pregnant mice, through increased proliferation and angiogenesis. In a SCD mouse model that recapitulates refractory SCD ulcers, we showed that the ulcers of post-partum SCD mice healed more quickly than those of virgin mice. This was associated with the recruitment of fetal cells in maternal wounds where they harbored markers of leukocytes and endothelial cells. In a retrospective cohort of SCD patients, using several parameters we found that SCD women who had ever had a baby had less of a burden related to leg ulcers compared to nulliparous women. Taken together, these results indicate that healing capacities of FMC are maintained long after delivery and may be exploited to promote wound healing in post-partum SCD patients.
Collapse
Affiliation(s)
- Mansour Alkobtawi
- Cutaneous Biology Lab, Institut Cochin, INSERM U1016, UMR 8104, Paris
| | - Maria Sbeih
- Cutaneous Biology Lab, Institut Cochin, INSERM U1016, UMR 8104, Paris
| | - Karim Souaid
- Department of Dermatology, Hôpital Cochin, AP-HP.Centre-Université Paris Cité, Paris, France; University Paris Cité, Faculté de Médecine Paris Centre Santé, Paris
| | - Qui Trung Ngô
- Cutaneous Biology Lab, Institut Cochin, INSERM U1016, UMR 8104, Paris
| | - Dany Nassar
- Cutaneous Biology Lab, Institut Cochin, INSERM U1016, UMR 8104, Paris, France; Department of Dermatology, Hôpital Cochin, AP-HP.Centre-Université Paris Cité, Paris, France; University Paris Cité, Faculté de Médecine Paris Centre Santé, Paris
| | - Hugo Arbes
- Institut de Biologie Intégrative de la Cellule, Genomic structure and Translation Lab, UMR_9198, CEA, CNRS, Université Paris-Saclay, Orsay
| | - Henri Guillet
- Department of Internal Medicine, Red Blood Cell Genetic Diseases Unit, Hôpital Mondor, AP-HP. Hôpitaux Universitaires Henri Mondor, Créteil
| | - Anoosha Habibi
- Department of Internal Medicine, Red Blood Cell Genetic Diseases Unit, Hôpital Mondor, AP-HP. Hôpitaux Universitaires Henri Mondor, Créteil
| | - Pablo Bartolucci
- Department of Internal Medicine, Red Blood Cell Genetic Diseases Unit, Hôpital Mondor, AP-HP. Hôpitaux Universitaires Henri Mondor, Créteil
| | - Mathieu Castela
- Cutaneous Biology Lab, Institut Cochin, INSERM U1016, UMR 8104, Paris
| | - Sélim Aractingi
- Cutaneous Biology Lab, Institut Cochin, INSERM U1016, UMR 8104, Paris, France; Department of Dermatology, Hôpital Cochin, AP-HP.Centre-Université Paris Cité, Paris, France; University Paris Cité, Faculté de Médecine Paris Centre Santé, Paris.
| | - Bénédicte Oulès
- Cutaneous Biology Lab, Institut Cochin, INSERM U1016, UMR 8104, Paris, France; Department of Dermatology, Hôpital Cochin, AP-HP.Centre-Université Paris Cité, Paris, France; University Paris Cité, Faculté de Médecine Paris Centre Santé, Paris
| |
Collapse
|
4
|
Sedov E, McCarthy J, Koren E, Fuchs Y. Fetomaternal microchimerism in tissue repair and tumor development. Dev Cell 2022; 57:1442-1452. [PMID: 35700729 DOI: 10.1016/j.devcel.2022.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/11/2022] [Accepted: 05/20/2022] [Indexed: 11/03/2022]
Abstract
In various placental mammals, the bidirectional exchange of cells during pregnancy can lead to the acquisition of genetically unique cells that can persist in both mother and child for decades. Over the years, it has become increasingly clear that this phenomenon, termed fetomaternal microchimerism may play key roles in a number of biological processes. In this perspective, we explore the concept of fetomaternal microchimerism and outline how fetal microchimeric cells are detected and immunologically tolerated within the maternal setting. Moreover, we discuss undertakings in the field that hint at the significant plasticity of fetal microchimeric cells and their potential roles in promoting maternal wound healing. Finally, we explore the multifaceted roles of fetal microchimeric cells in cancer development and progression. A deeper understanding of fetomaternal chimerism in healthy and diseased states will be key toward developing more efficient anti-cancer treatments and regenerative therapies.
Collapse
Affiliation(s)
- Egor Sedov
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa 3200003, Israel; Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa 3200003, Israel; Technion Integrated Cancer Center, Technion Israel Institute of Technology, Haifa 3200003, Israel
| | - Jordan McCarthy
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa 3200003, Israel; Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa 3200003, Israel; Technion Integrated Cancer Center, Technion Israel Institute of Technology, Haifa 3200003, Israel
| | - Elle Koren
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa 3200003, Israel; Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa 3200003, Israel; Technion Integrated Cancer Center, Technion Israel Institute of Technology, Haifa 3200003, Israel
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa 3200003, Israel; Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa 3200003, Israel; Technion Integrated Cancer Center, Technion Israel Institute of Technology, Haifa 3200003, Israel.
| |
Collapse
|
5
|
Jahanbani Y, Shafiee S, Davaran S, Roshangar L, Ahmadian E, Eftekhari A, Dolati S, Yousefi M. Stem cells technology as a platform for generating reproductive system organoids and treatment of infertility-related diseases. Cell Biol Int 2022; 46:512-522. [PMID: 34918417 DOI: 10.1002/cbin.11747] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/14/2021] [Indexed: 02/06/2023]
Abstract
In recent years, stem cells have known as a helpful biological tool for the accurate diagnosis, treatment and recognition of diseases. Using stem cells as biomarkers have presented high potential in the early detection of many diseases. Another advancement in stem cell technology includes stem cell derived organoids model that could be a promising platform for diagnosis and modeling different diseases. Furthermore, therapeutic capabilities of stem cell therapy have increased hope in the face of different disability managements. All of these technologies are also widely used in reproductive related diseases especially in today's world that many couples encounter infertility problems. However, with the aid of numerous improvements in the treatment of infertility, over 80% of couples who dreamed of having children could now have children. Due to the fact that infertility has many negative effects on personal and social lives of young couples, many researchers have focused on the treatment of male and female reproductive system abnormalities with different types of stem cells, including embryonic stem cells, bone marrow mesenchymal stem cells (MSCs), and umbilical cord-derived MSCs. Also, design and formation of reproductive system organoids provide a fascinating window into disease modeling, drug screening, personalized therapy, and regeneration medicine. Utilizing these techniques to study, model and treat the infertility-related diseases has drawn attention of many scientists. This review explains different applications of stem cells in generating reproductive system organoids and stem cell-based therapies for male and female infertility related diseases treatment.
Collapse
Affiliation(s)
- Yalda Jahanbani
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samira Shafiee
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soodabeh Davaran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aziz Eftekhari
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Russian Institute for Advanced Study, Moscow State Pedagogical University, Moscow, Russian Federation
| | - Sanam Dolati
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Engels G, Döhler B, Tönshoff B, Oh J, Kruchen A, Müller I, Süsal C. Maternal versus paternal living kidney transplant donation is associated with lower rejection in young pediatric recipients: A Collaborative Transplant Study report. Pediatr Transplant 2022; 26:e14154. [PMID: 34612565 DOI: 10.1111/petr.14154] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/02/2021] [Accepted: 09/13/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Approximately 1700 children per year with end-stage kidney disease undergo kidney transplantation in Europe and the United States of America; 30%-50% are living donor kidney transplantations. There may be immunological differences between paternal and maternal donors due to transplacental exchange of cells between the mother and fetus during pregnancy leading to microchimerism. We investigated whether the outcome of living-related kidney transplantation in young children is different after maternal compared with paternal organ donation. METHODS Using the international Collaborative Transplant Study (CTS) database, we analyzed epidemiological data of 7247 children and adolescents aged <18 years who had received a kidney transplant from either mother or father. Risk of treated rejection episodes and death-censored graft failure were computed using the Kaplan-Meier method and multivariable Cox regression. RESULTS In the recipient age group 1-4 years, the rate of treated rejection episodes in recipients of kidneys from maternal donors (N = 195) during the first 2 years post-transplant was significantly lower (hazard ratio HR = 0.47, p = .004) than in patients receiving kidneys from paternal donors (N = 179). This association between donor sex and risk of treated rejections was not observed in children aged 5-9 years. The 5-year death-censored graft survival in children aged 1-4 years with a maternal or paternal donor was comparable. CONCLUSIONS Maternal kidney donation in young pediatric renal transplant recipients is associated with an approximately 50% lower rate of treated rejection than paternal kidney donation. Whether this phenomenon is due to maternal microchimerism-induced donor-specific hyporesponsiveness must be evaluated in prospective mechanistic studies.
Collapse
Affiliation(s)
- Geraldine Engels
- Department of Pediatrics, University Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Pediatrics, University of Würzburg, Würzburg, Germany
| | - Bernd Döhler
- Institute of Immunology, Transplantation Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Burkhard Tönshoff
- Department of Pediatrics I, University Children's Hospital, University of Heidelberg, Heidelberg, Germany
| | - Jun Oh
- Department of Pediatrics, University Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Kruchen
- Division of Pediatric Stem Cell Transplantation and Immunology, Department of Pediatric Hematology and Oncology, University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| | - Ingo Müller
- Division of Pediatric Stem Cell Transplantation and Immunology, Department of Pediatric Hematology and Oncology, University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| | - Caner Süsal
- Institute of Immunology, Transplantation Immunology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
7
|
Sabbatinelli G, Fantasia D, Palka C, Morizio E, Alfonsi M, Calabrese G. Isolation and Enrichment of Circulating Fetal Cells for NIPD: An Overview. Diagnostics (Basel) 2021; 11:diagnostics11122239. [PMID: 34943476 PMCID: PMC8700692 DOI: 10.3390/diagnostics11122239] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 11/26/2021] [Accepted: 11/28/2021] [Indexed: 11/17/2022] Open
Abstract
Prenatal diagnosis plays a crucial role in clinical genetics. Non-invasive prenatal diagnosis using fetal cells circulating in maternal peripheral blood has become the goal of prenatal diagnosis, to obtain complete fetal genetic information and avoid risks to mother and fetus. The development of high-efficiency separation technologies is necessary to obtain the scarce fetal cells from the maternal circulation. Over the years, multiple approaches have been applied, including choice of the ideal cell targets, different cell recovering technologies, and refined cell isolation yield procedures. In order to provide a useful tool and to give insights about limitations and advantages of the technologies available today, we review the genetic research on the creation and validation of non-invasive prenatal diagnostic testing protocols based on the rare and labile circulating fetal cells during pregnancy.
Collapse
Affiliation(s)
- Giulia Sabbatinelli
- Dipartimento di Neuroscienze, Imaging & Scienze Cliniche, Scuola Superiore G. D’Annunzio, University of Chieti, 66100 Chieti, Italy;
| | - Donatella Fantasia
- UOSD Genetica Oncoematologica, Dipartimento di Oncologico-Ematologico, Ospedale Spirito Santo, ASL Pescara, 65124 Pescara, Italy;
| | - Chiara Palka
- UOC Genetica Medica, Ospedale S.S. Annunziata, ASL2 Chieti, 66100 Chieti, Italy; (C.P.); (M.A.)
| | - Elisena Morizio
- Genetica Medica, Dipartimento di Tecnologie Avanzate in Medicina e Odontoiatria, School of Medicine, University of Chieti, 66100 Chieti, Italy;
| | - Melissa Alfonsi
- UOC Genetica Medica, Ospedale S.S. Annunziata, ASL2 Chieti, 66100 Chieti, Italy; (C.P.); (M.A.)
| | - Giuseppe Calabrese
- UOSD Genetica Oncoematologica, Dipartimento di Oncologico-Ematologico, Ospedale Spirito Santo, ASL Pescara, 65124 Pescara, Italy;
- Genetica Medica, Dipartimento di Tecnologie Avanzate in Medicina e Odontoiatria, School of Medicine, University of Chieti, 66100 Chieti, Italy;
- Correspondence:
| |
Collapse
|
8
|
Rosner M, Kolbe T, Hengstschläger M. Fetomaternal microchimerism and genetic diagnosis: On the origins of fetal cells and cell-free fetal DNA in the pregnant woman. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2021; 788:108399. [PMID: 34893150 DOI: 10.1016/j.mrrev.2021.108399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/11/2021] [Accepted: 11/14/2021] [Indexed: 06/14/2023]
Abstract
During pregnancy several types of fetal cells and fetal stem cells, including pregnancy-associated progenitor cells (PAPCs), traffic into the maternal circulation. Whereas they also migrate to various maternal organs and adopt the phenotype of the target tissues to contribute to regenerative processes, fetal cells also play a role in the pathogenesis of maternal diseases. In addition, cell-free fetal DNA (cffDNA) is detectable in the plasma of pregnant women. Together they constitute the well-known phenomenon of fetomaternal microchimerism, which inspired the concept of non-invasive prenatal testing (NIPT) using maternal blood. An in-depth knowledge concerning the origins of these fetal cells and cffDNA allows a more comprehensive understanding of the biological relevance of fetomaternal microchimerism and has implications for the ongoing expansion of resultant clinical applications.
Collapse
Affiliation(s)
- Margit Rosner
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Thomas Kolbe
- Biomodels Austria, University of Veterinary Medicine Vienna, Vienna, Austria; Department IFA Tulln, University of Natural Resources and Life Sciences, Tulln, Austria
| | - Markus Hengstschläger
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
9
|
Bianchi DW, Khosrotehrani K, Way SS, MacKenzie TC, Bajema I, O'Donoghue K. Forever Connected: The Lifelong Biological Consequences of Fetomaternal and Maternofetal Microchimerism. Clin Chem 2020; 67:351-362. [PMID: 33417673 PMCID: PMC10072000 DOI: 10.1093/clinchem/hvaa304] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 10/28/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND Originally studied as a mechanism to understand eclampsia-related deaths during pregnancy, fetal cells in maternal blood have more recently garnered attention as a noninvasive source of fetal material for prenatal testing. In the 21st century, however, intact fetal cells have been largely supplanted by circulating cell-free placental DNA for aneuploidy screening. Instead, interest has pivoted to the ways in which fetal cells influence maternal biology. In parallel, an increasing appreciation of the consequences of maternal cells in the developing fetus has occurred. CONTENT In this review, we highlight the potential clinical applications and functional consequences of the bidirectional trafficking of intact cells between a pregnant woman and her fetus. Fetal cells play a potential role in the pathogenesis of maternal disease and tissue repair. Maternal cells play an essential role in educating the fetal immune system and as a factor in transplant acceptance. Naturally occurring maternal microchimerism is also being explored as a source of hematopoietic stem cells for transplant in fetal hematopoietic disorders. SUMMARY Future investigations in humans need to include complete pregnancy histories to understand maternal health and transplant success or failure. Animal models are useful to understand the mechanisms underlying fetal wound healing and/or repair associated with maternal injury and inflammation. The lifelong consequences of the exchange of cells between a mother and her child are profound and have many applications in development, health, and disease. This intricate exchange of genetically foreign cells creates a permanent connection that contributes to the survival of both individuals.
Collapse
Affiliation(s)
- Diana W Bianchi
- National Human Genome Research Institute and Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Kiarash Khosrotehrani
- Experimental Dermatology Group, The University of Queensland, UQ Diamantina Institute, Brisbane, Queensland, Australia
| | - Sing Sing Way
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tippi C MacKenzie
- Center for Maternal-Fetal Precision Medicine and the Department of Surgery, University of California, San Francisco, CA, USA
| | - Ingeborg Bajema
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Keelin O'Donoghue
- Irish Centre for Maternal and Child Health (INFANT), University College Cork, Cork, Ireland
| |
Collapse
|
10
|
Koh B, Sulaiman N, Fauzi MB, Law JX, Ng MH, Idrus RBH, Yazid MD. Three dimensional microcarrier system in mesenchymal stem cell culture: a systematic review. Cell Biosci 2020; 10:75. [PMID: 32518618 PMCID: PMC7271456 DOI: 10.1186/s13578-020-00438-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 05/27/2020] [Indexed: 01/09/2023] Open
Abstract
Stem cell-based regenerative medicine is a promising approach for tissue reconstruction. However, a large number of cells are needed in a typical clinical study, where conventional monolayer cultures might pose a limitation for scale-up. The purpose of this review was to systematically assess the application of microcarriers in Mesenchymal Stem Cell cultures. A comprehensive search was conducted in Medline via Ebscohost, Pubmed, and Scopus, and relevant studies published between 2015 and 2019 were selected. The literature search identified 53 related studies, but only 14 articles met the inclusion criteria. These include 7 utilised commercially available microcarriers, while the rest were formulated based on different surface characteristics, all of which are discussed in this review. Current applications of microcarriers were focused on MSC expansion and induction of MSCs into different lineages. These studies demonstrated that MSCs could proliferate in a microcarrier culture system in-fold compared to monolayer cultures, and the culture system could simulate a three-dimensional environment which induces cell differentiation. However, detailed studies are still required before this system were to be adapted into the scale of GMP manufacturing.
Collapse
Affiliation(s)
- Benson Koh
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Nadiah Sulaiman
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Mh Busra Fauzi
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Jia Xian Law
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Min Hwei Ng
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Ruszymah Bt Hj Idrus
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia.,Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Muhammad Dain Yazid
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia
| |
Collapse
|
11
|
Johnson BN, Ehli EA, Davies GE, Boomsma DI. Chimerism in health and potential implications on behavior: A systematic review. Am J Med Genet A 2020; 182:1513-1529. [PMID: 32212323 DOI: 10.1002/ajmg.a.61565] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 02/03/2020] [Accepted: 03/02/2020] [Indexed: 12/12/2022]
Abstract
In this review, we focus on the phenomenon of chimerism and especially microchimerism as one of the currently underexplored explanations for differences in health and behavior. Chimerism is an amalgamation of cells from two or more unique zygotes within a single organism, with microchimerism defined by a minor cell population of <1%. This article first presents an overview of the primary techniques employed to detect and quantify the presence of microchimerism and then reviews empirical studies of chimerism in mammals including primates and humans. In women, male microchimerism, a condition suggested to be the result of fetomaternal exchange in utero, is relatively easily detected by polymerase chain reaction molecular techniques targeting Y-chromosomal markers. Consequently, studies of chimerism in human diseases have largely focused on diseases with a predilection for females including autoimmune diseases, and female cancers. We detail studies of chimerism in human diseases and also discuss some potential implications in behavior. Understanding the prevalence of chimerism and the associated health outcomes will provide invaluable knowledge of human biology and guide novel approaches for treating diseases.
Collapse
Affiliation(s)
- Brandon N Johnson
- Avera Institute for Human Genetics, Avera McKennan Hospital and University Health Center, Sioux Falls, South Dakota, USA
| | - Erik A Ehli
- Avera Institute for Human Genetics, Avera McKennan Hospital and University Health Center, Sioux Falls, South Dakota, USA
| | - Gareth E Davies
- Avera Institute for Human Genetics, Avera McKennan Hospital and University Health Center, Sioux Falls, South Dakota, USA
| | - Dorret I Boomsma
- Netherlands Twin Register, Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Cismaru CA, Pop L, Berindan-Neagoe I. Incognito: Are Microchimeric Fetal Stem Cells that Cross Placental Barrier Real Emissaries of Peace? Stem Cell Rev Rep 2018; 14:632-641. [PMID: 29948753 DOI: 10.1007/s12015-018-9834-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Chimerism occurs naturaly throughout gestation and can also occur as a consequence of transfusion and transplantation therapy. It consists of the acquisition and long-term persistence of a genetically distinct population of allogenic cells inside another organism. Previous reports have suggested that feto-maternal microchimerism could exert a beneficial effect on the treatment of hematological and solid tumors in patients treated by PBSCT. In this review we report the mechanism of transplacental fetal stem cell trafficking during pregnancy and the effect of their long-term persistence on autoimmunity, GVHD, PBSCT, cancer and stem cell treatment.
Collapse
Affiliation(s)
- Cosmin Andrei Cismaru
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, 23 Gh. Marinescu street, 400337, Cluj-Napoca, Romania.
| | - Laura Pop
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, 23 Gh. Marinescu street, 400337, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, 23 Gh. Marinescu street, 400337, Cluj-Napoca, Romania
| |
Collapse
|
13
|
Ansari AS, Yazid MD, Sainik NQAV, Razali RA, Saim AB, Idrus RBH. Osteogenic Induction of Wharton's Jelly-Derived Mesenchymal Stem Cell for Bone Regeneration: A Systematic Review. Stem Cells Int 2018; 2018:2406462. [PMID: 30534156 PMCID: PMC6252214 DOI: 10.1155/2018/2406462] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 07/27/2018] [Accepted: 09/03/2018] [Indexed: 12/13/2022] Open
Abstract
Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) are emerging as a promising source for bone regeneration in the treatment of bone defects. Previous studies have reported the ability of WJ-MSCs to be induced into the osteogenic lineage. The purpose of this review was to systematically assess the potential of WJ-MSC differentiation into the osteogenic lineage. A comprehensive search was conducted in Medline via Ebscohost and Scopus, where relevant studies published between 1961 and 2018 were selected. The main inclusion criteria were that articles must be primary studies published in English evaluating osteogenic induction of WJ-MSCs. The literature search identified 92 related articles, but only 18 articles met the inclusion criteria. These include two animal studies, three articles containing both in vitro and in vivo assessments, and 13 articles on in vitro studies, all of which are discussed in this review. There were two types of osteogenic induction used in these studies, either chemical or physical. The studies demonstrate that WJ-MSCs are able to differentiate into osteogenic lineage and promote osteogenesis. In light of these observations, it is suggested that WJ-MSCs can be a potential source of stem cells for osteogenic induction, as an alternative to bone marrow-derived mesenchymal stem cells.
Collapse
Affiliation(s)
- Ayu Suraya Ansari
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Muhammad Dain Yazid
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Nur Qisya Afifah Veronica Sainik
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Rabiatul Adawiyah Razali
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Aminuddin Bin Saim
- Ear, Nose & Throat Consultant Clinic, Ampang Puteri Specialist Hospital, 68000 Ampang, Selangor, Malaysia
| | - Ruszymah Bt Hj Idrus
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
14
|
Ståhlberg A, El-Heliebi A, Sedlmayr P, Kroneis T. Unravelling the biological secrets of microchimerism by single-cell analysis. Brief Funct Genomics 2018; 17:255-264. [PMID: 29028900 PMCID: PMC6063264 DOI: 10.1093/bfgp/elx027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The presence of microchimeric cells is known for >100 years and well documented since decades. Earlier, microchimeric cells were mainly used for cell-based non-invasive prenatal diagnostics during early pregnancy. Microchimeric cells are also present beyond delivery and are associated to various autoimmune diseases, tissue repair, cancer and immune tolerance. All these findings were based on low complexity studies and occasionally accompanied by artefacts not allowing the biological functions of microchimerism to be determined. However, with the recent developments in single-cell analysis, new means to identify and characterize microchimeric cells are available. Cell labelling techniques in combination with single-cell analysis provide a new toolbox to decipher the biology of microchimeric cells at molecular and cellular level. In this review, we discuss how recent developments in single-cell analysis can be applied to determine the role and function of microchimeric cells.
Collapse
Affiliation(s)
- Anders Ståhlberg
- Sahlgrenska Cancer Center, Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 1F, Gothenburg, Sweden
| | - Amin El-Heliebi
- Institute of Cell Biology, Histology & Embryology, Medical University of Graz, Harrachgasse 21, Graz, Austria
| | - Peter Sedlmayr
- Institute of Cell Biology, Histology & Embryology, Medical University of Graz, Harrachgasse 21, Graz, Austria
| | - Thomas Kroneis
- Sahlgrenska Cancer Center, Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 1F, Gothenburg, Sweden
- Institute of Cell Biology, Histology & Embryology, Medical University of Graz, Harrachgasse 21, Graz, Austria
| |
Collapse
|
15
|
Zhong JF, Weiner LP. Role of Fetal Stem Cells in Maternal Tissue Regeneration. GENE REGULATION AND SYSTEMS BIOLOGY 2017. [DOI: 10.1177/117762500700100011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Microchimerism refers to the status of harboring cells from another individual at low levels. It is well known that cells traffic bidirectionally between fetus and mother during pregnancy. This situation resembles a naturally occurring long lasting fetal stem cell transplantation. The fetus acts as the donor and the mother acts as the recipient. To study the role of microchimerism in tissue regeneration, we constructed a murine microchimerism model with wild type C57BL/6J female mice carrying progenies which expressed green fluorescent proteins (GFP). Our data indicated that skin injuries in the female mice during pregnancy increased microchimerism of GFP expressing cells from the GFP transgenic progenies. The GFP positive cells also appeared at the site of spinal cord where injury occurred during pregnancy. Our study suggests that the amount of fetal cells in maternal mice significantly increased if injuries occurred during pregnancy. Fetal stem cells appear to respond to maternal injury signals and may play a role in maternal tissue regeneration during pregnancy.
Collapse
Affiliation(s)
- Jiang F. Zhong
- The Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, U.S.A
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, U.S.A
| | - Leslie P. Weiner
- The Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, U.S.A
| |
Collapse
|
16
|
Aqmasheh S, Shamsasanjan K, Akbarzadehlaleh P, Pashoutan Sarvar D, Timari H. Effects of Mesenchymal Stem Cell Derivatives on Hematopoiesis and Hematopoietic Stem Cells. Adv Pharm Bull 2017; 7:165-177. [PMID: 28761818 PMCID: PMC5527230 DOI: 10.15171/apb.2017.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 04/08/2017] [Accepted: 04/18/2017] [Indexed: 12/11/2022] Open
Abstract
Hematopoiesis is a balance among quiescence, self-renewal, proliferation, and differentiation, which is believed to be firmly adjusted through interactions between hematopoietic stem and progenitor cells (HSPCs) with the microenvironment. This microenvironment is derived from a common progenitor of mesenchymal origin and its signals should be capable of regulating the cellular memory of transcriptional situation and lead to an exchange of stem cell genes expression. Mesenchymal stem cells (MSCs) have self-renewal and differentiation capacity into tissues of mesodermal origin, and these cells can support hematopoiesis through release various molecules that play a crucial role in migration, homing, self-renewal, proliferation, and differentiation of HSPCs. Studies on the effects of MSCs on HSPC differentiation can develop modern solutions in the treatment of patients with hematologic disorders for more effective Bone Marrow (BM) transplantation in the near future. However, considerable challenges remain on realization of how paracrine mechanisms of MSCs act on the target tissues, and how to design a therapeutic regimen with various paracrine factors in order to achieve optimal results for tissue conservation and regeneration. The aim of this review is to characterize and consider the related aspects of the ability of MSCs secretome in protection of hematopoiesis.
Collapse
Affiliation(s)
- Sara Aqmasheh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasanjan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Akbarzadehlaleh
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hamze Timari
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Castela M, Nassar D, Sbeih M, Jachiet M, Wang Z, Aractingi S. Ccl2/Ccr2 signalling recruits a distinct fetal microchimeric population that rescues delayed maternal wound healing. Nat Commun 2017; 8:15463. [PMID: 28516946 PMCID: PMC5477505 DOI: 10.1038/ncomms15463] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 03/31/2017] [Indexed: 12/20/2022] Open
Abstract
Foetal microchimeric cells (FMCs) traffic into maternal circulation during pregnancy and persist for decades after delivery. Upon maternal injury, FMCs migrate to affected sites where they participate in tissue healing. However, the specific signals regulating the trafficking of FMCs to injury sites had to be identified. Here we report that, in mice, a subset of FMCs implicated in tissue repair displays CD11b+ CD34+ CD31+ phenotype and highly express C-C chemokine receptor 2 (Ccr2). The Ccr2 ligand chemokine ligand 2 (Ccl2) enhances the recruitment of FMCs to maternal wounds where these cells transdifferentiate into endothelial cells and stimulate angiogenesis through Cxcl1 secretion. Ccl2 administration improves delayed maternal wound healing in pregnant and postpartum mice but never in virgin ones. This role of Ccl2/Ccr2 signalling opens new strategies for tissue repair through natural stem cell therapy, a concept that can be later applied to other types of maternal diseases.
Collapse
Affiliation(s)
- Mathieu Castela
- INSERM UMRS_938, Saint-Antoine Research Center, 27, rue de Chaligny, Paris 75012, France.,UPMC Université Paris 6, 4, place Jussier, Paris 75005, France
| | - Dany Nassar
- Université Paris 5 Descartes, 12, rue de l'Ecole de Médecine, Paris 75006, France.,Department of Dermatology, American University of Beirut, Riad EI Soph, Beinut 11072020, Lebanon
| | - Maria Sbeih
- INSERM UMRS_938, Saint-Antoine Research Center, 27, rue de Chaligny, Paris 75012, France.,UPMC Université Paris 6, 4, place Jussier, Paris 75005, France
| | - Marie Jachiet
- INSERM UMRS_938, Saint-Antoine Research Center, 27, rue de Chaligny, Paris 75012, France.,UPMC Université Paris 6, 4, place Jussier, Paris 75005, France.,Université Paris 5 Descartes, 12, rue de l'Ecole de Médecine, Paris 75006, France
| | - Zhe Wang
- INSERM UMRS_938, Saint-Antoine Research Center, 27, rue de Chaligny, Paris 75012, France
| | - Selim Aractingi
- INSERM UMRS_938, Saint-Antoine Research Center, 27, rue de Chaligny, Paris 75012, France.,Université Paris 5 Descartes, 12, rue de l'Ecole de Médecine, Paris 75006, France.,Department of Dermatology, Hôpital Cochin, AP-HP, 89, rue d'Assas, Paris 75006, France
| |
Collapse
|
18
|
Ebrahim NA, Leach L. Transendothelial migration of human umbilical mesenchymal stem cells across uterine endothelial monolayers: Junctional dynamics and putative mechanisms. Placenta 2016; 48:87-98. [DOI: 10.1016/j.placenta.2016.10.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 10/19/2016] [Accepted: 10/20/2016] [Indexed: 11/28/2022]
|
19
|
Abstract
The ischemia-induced death of cardiomyocytes results in scar formation and reduced contractility of the ventricle. Several preclinical and clinical studies have supported the notion that cell therapy may be used for cardiac regeneration. Most attempts for cardiomyoplasty have considered the bone marrow as the source of the “repair stem cell(s),” assuming that the hematopoietic stem cell can do the work. However, bone marrow is also the residence of other progenitor cells, including mesenchymal stem cells (MSCs). Since 1995 it has been known that under in vitro conditions, MSCs differentiate into cells exhibiting features of cardiomyocytes. This pioneer work was followed by many preclinical studies that revealed that ex vivo expanded, bone marrow–derived MSCs may represent another option for cardiac regeneration. In this work, we review evidence and new prospects that support the use of MSCs in cardiomyoplasty.
Collapse
Affiliation(s)
- José J Minguell
- Laboratorio de Trasplante de Médula Osea, Clínica Las Condes, Lo Fontecilla 441, Las Condes, Santiago, Chile.
| | | |
Collapse
|
20
|
Valenti MT, Mori A, Malerba G, Dalle Carbonare L. Mesenchymal stem cells: A new diagnostic tool? World J Stem Cells 2015; 7:789-792. [PMID: 26131309 PMCID: PMC4478625 DOI: 10.4252/wjsc.v7.i5.789] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 03/27/2015] [Accepted: 04/30/2015] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are progenitor cells capable of self-renewal that can differentiate in multiple tissues and, under specific and standardized culture conditions, expand in vitro with little phenotypic alterations. In recent years, preclinical and clinical studies have focused on MSC analysis and understanding the potential use of these cells as a therapy in a wide range of pathologies, and many applications have been tested. Clinical trials using MSCs have been performed (e.g., for cardiac events, stroke, multiple sclerosis, blood diseases, auto-immune disorders, ischemia, and articular cartilage and bone pathologies), and for many genetic diseases, these cells are considered an important resource. Considering of the biology of MSCs, these cells may also be useful tools for understanding the physiopathology of different diseases, and they can be used to develop specific biomarkers for a broad range of diseases. In this editorial, we discuss the literature related to the use of MSCs for diagnostic applications and we suggest new technologies to improve their employment.
Collapse
|
21
|
Habibollah S, Forraz N, McGuckin CP. Application of Umbilical Cord and Cord Blood as Alternative Modes for Liver Therapy. Regen Med 2015. [DOI: 10.1007/978-1-4471-6542-2_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
22
|
Bhattacharya N, Das SP, Sengupta DB, Chowdhury P, Chowdhury D, Das K, Das S, Maity N, Bhattacharya R, Sengupta D, Aikat A, Basu D, Chaudhuri S, Rakshit T, Bhattacharya A, Bhattacharya SK, Majumder U, Chakraborty B, Chaudhuri S, Law S, Tripathi SK, Basu N, Banerjee SK, Malakar D, Choudhuri S. Chronic Burn Ulceration of the Skin and the Potential of Amniotic Membrane-Based Therapy. Regen Med 2015. [DOI: 10.1007/978-1-4471-6542-2_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
23
|
|
24
|
Heazlewood CF, Sherrell H, Ryan J, Atkinson K, Wells CA, Fisk NM. High incidence of contaminating maternal cell overgrowth in human placental mesenchymal stem/stromal cell cultures: a systematic review. Stem Cells Transl Med 2014; 3:1305-11. [PMID: 25154781 DOI: 10.5966/sctm.2014-0051] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Placenta is a readily accessible translationally advantageous source of mesenchymal stem/stromal cells (MSCs) currently used in cryobanking and clinical trials. MSCs cultured from human chorion have been widely assumed to be fetal in origin, despite evidence that placental MSCs may be contaminated with maternal cells, resulting in entirely maternally derived MSC cultures. To document the frequency and determinants of maternal cell contamination in chorionic MSCs, we undertook a PRISMA-compliant systematic review of publications in the PubMed, Medline, and Embase databases (January 2000 to July 2013) on placental and/or chorionic MSCs from uncomplicated pregnancies. Of 147 studies, only 26 (18%) investigated fetal and/or maternal cell origin. After excluding studies that did not satisfy minimal MSC criteria, 7 of 15 informative studies documented MSC cultures as entirely fetal, a further 7 studies reported cultured human chorionic MSC populations to be either maternal (n=6) or mixed (n=1), whereas 1 study separately cultured pure fetal and pure maternal MSC from the same placenta. Maternal cell contamination was associated with term and chorionic membrane samples and greater passage number but was still present in 30% of studies of chorionic villous MSCs. Although most studies assume fetal origin for MSCs sourced from chorion, this systematic review documents a high incidence of maternal-origin MSC populations in placental MSC cultures. Given that fetal MSCs have more primitive properties than adult MSCs, our findings have implications for clinical trials in which knowledge of donor and tissue source is pivotal. We recommend sensitive methods to quantitate the source and purity of placental MSCs.
Collapse
Affiliation(s)
- Celena F Heazlewood
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia; University of Queensland Centre for Clinical Research, University of Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology at the Translational Research Institute, and Centre for Advanced Prenatal Care, Royal Brisbane & Women's Hospital, Brisbane, Queensland, Australia; Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Helen Sherrell
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia; University of Queensland Centre for Clinical Research, University of Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology at the Translational Research Institute, and Centre for Advanced Prenatal Care, Royal Brisbane & Women's Hospital, Brisbane, Queensland, Australia; Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Jennifer Ryan
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia; University of Queensland Centre for Clinical Research, University of Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology at the Translational Research Institute, and Centre for Advanced Prenatal Care, Royal Brisbane & Women's Hospital, Brisbane, Queensland, Australia; Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Kerry Atkinson
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia; University of Queensland Centre for Clinical Research, University of Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology at the Translational Research Institute, and Centre for Advanced Prenatal Care, Royal Brisbane & Women's Hospital, Brisbane, Queensland, Australia; Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Christine A Wells
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia; University of Queensland Centre for Clinical Research, University of Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology at the Translational Research Institute, and Centre for Advanced Prenatal Care, Royal Brisbane & Women's Hospital, Brisbane, Queensland, Australia; Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Nicholas M Fisk
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia; University of Queensland Centre for Clinical Research, University of Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology at the Translational Research Institute, and Centre for Advanced Prenatal Care, Royal Brisbane & Women's Hospital, Brisbane, Queensland, Australia; Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
25
|
Tanabe S. Role of mesenchymal stem cells in cell life and their signaling. World J Stem Cells 2014; 6:24-32. [PMID: 24567785 PMCID: PMC3927011 DOI: 10.4252/wjsc.v6.i1.24] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 11/18/2013] [Accepted: 12/13/2013] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have various roles in the body and cellular environment, and the cellular phenotypes of MSCs changes in different conditions. MSCs support the maintenance of other cells, and the capacity of MSCs to differentiate into several cell types makes the cells unique and full of possibilities. The involvement of MSCs in the epithelial-mesenchymal transition is an important property of these cells. In this review, the role of MSCs in cell life, including their application in therapy, is first described, and the signaling mechanism of MSCs is investigated for a further understanding of these cells.
Collapse
|
26
|
Biphasic recruitment of microchimeric fetal mesenchymal cells in fibrosis following acute kidney injury. Kidney Int 2013; 85:600-10. [PMID: 24304884 DOI: 10.1038/ki.2013.459] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 09/03/2013] [Accepted: 09/05/2013] [Indexed: 12/19/2022]
Abstract
Fetal microchimeric cells (FMCs) enter the maternal circulation and persist in tissue for decades. They have capacity to home to injured maternal tissue and differentiate along that tissue's lineage. This raises the question of the origin(s) of cells transferred to the mother during pregnancy. FMCs with a mesenchymal phenotype have been documented in several studies, which makes mesenchymal stem cells an attractive explanation for their broad plasticity. Here we assessed the recruitment and mesenchymal lineage contribution of FMCs in response to acute kidney fibrosis induced by aristolochic acid injection. Serial in vivo bioluminescence imaging revealed a biphasic recruitment of active collagen-producing FMCs during the repair process of injured kidney in post-partum wild-type mothers that had delivered transgenic pups expressing luciferase under the collagen type I-promoter. The presence of FMCs long-term post injury (day 60) was associated with profibrotic molecules (TGF-β/CTGF), serum urea levels, and collagen deposition. Immunostaining confirmed FMCs at short term (day 15) using post-partum wild-type mothers that had delivered green fluorescent protein-positive pups and suggested a mainly hematopoietic phenotype. We conclude that there is biphasic recruitment to, and activity of, FMCs at the injury site. Moreover, we identified five types of FMC, implicating them all in the reparative process at different stages of induced renal interstitial fibrosis.
Collapse
|
27
|
Hromadnikova I, Kotlabova K, Pirkova P, Libalova P, Vernerova Z, Svoboda B, Kucera E. The occurrence of fetal microchimeric cells in endometrial tissues is a very common phenomenon in benign uterine disorders, and the lower prevalence of fetal microchimerism is associated with better uterine cancer prognoses. DNA Cell Biol 2013; 33:40-8. [PMID: 24283364 DOI: 10.1089/dna.2013.2125] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
This is the first study carried out to describe the role of fetal microchimerism (FM) in the pathogenesis of uterine cancer. The prevalence and concentration of male fetal microchimeric cells (FMCs) were examined in endometrial tissues in relation to subtypes of uterine cancer, and the histological grade and stage of the tumor. FM occurrence was analyzed in relation to risk factors, including hypertension, obesity, type 2 diabetes, dyslipidemia, age at cancer diagnosis, and patient pregnancy history. The prevalence and concentration of FMCs were examined in endometrial tissues using real-time polymerase chain reaction, SRY and β-globin sequences as markers for male fetal FMCs and total DNA. The studied group involved 47 type 1 endometrial cancers, 28 type 2 endometrial cancers, and 41 benign uterine diseases. While the prevalence of FM was decreased only in type 1 endometrial cancer, compared with benign uterine disorders (38.3% vs.70.7%; odds ratio [OR]=0.257, 95% confidence interval [CI]: 0.105 to 0.628, p=0.003), FMC concentrations did not differ within examined groups. The lower FM prevalence was detected in low-grade (grade 1 and grade 2) endometrioid cancer (38.3% vs. 70.7%, OR=0.256, 95% CI: 0.105 to 0.627, p=0.003) and in FIGO 1 tumors (40.7% vs. 70.7%, OR=0.285, 95% CI: 0.120 to 0.675, p=0.004). No correlation between FM prevalence or FMC concentrations and risk factors was demonstrated. A lower prevalence of male FM seemed to be associated with better prognoses in uterine cancer based on tumor subtype, histological grade, and stage of the tumor.
Collapse
Affiliation(s)
- Ilona Hromadnikova
- 1 Department of Molecular Biology and Cell Pathology, Third Faculty of Medicine, Charles University in Prague , Prague, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
28
|
Duke CMP, Taylor HS. Stem cells and the reproductive system: historical perspective and future directions. Maturitas 2013; 76:284-9. [PMID: 24144960 DOI: 10.1016/j.maturitas.2013.08.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 08/29/2013] [Indexed: 12/20/2022]
Abstract
Recent findings in stem cell biology have presented new perspectives and opportunities for the treatment of reproductive disease. In a departure from the long held dogma of embryologically fixed numbers of oocytes, current literature suggests that human ovaries contain stem cells which form new oocytes even in adulthood and that these stem cells can be cultured in vitro to develop into mature oocytes. These findings have provided new hope and broader options for fertility preservation. Evidence of endometrial regeneration by bone marrow stem cells in endometrial tissue of women who received bone marrow transplant highlight potential for the novel treatments of uterine disorders and supports new theories for the etiology of endometriosis - ectopic transdifferentiation of stem cells. Further, endometrial derived stem cells have been demonstrated to be useful in the treatment of several chronic and often debilitating diseases, including Parkinson's Disease and Diabetes. Other cells that may present future therapeutic benefits for a myriad of disease states include placental and fetal cells which enter maternal circulation during pregnancy and can later promote parenchymal regeneration in maternal tissue. These findings highlight novel functions of the uterus and ovaries. They demonstrate that the uterus is a dynamic organ permeable to fetal stem cells capable of transdifferentiation as well as a renewable source of multipotent stem cells. While we still have much to understand about stem cells, their potential applications in reproductive biology and medicine are countless.
Collapse
Affiliation(s)
- Cindy M P Duke
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
29
|
Seppanen E, Fisk NM, Khosrotehrani K. Pregnancy-acquired fetal progenitor cells. J Reprod Immunol 2013; 97:27-35. [PMID: 23432869 DOI: 10.1016/j.jri.2012.08.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 08/23/2012] [Accepted: 08/23/2012] [Indexed: 12/11/2022]
Abstract
The transfer and persistence of fetal progenitor cells into the mother throughout pregnancy has sparked considerable interest as a trafficking stem cell and immunological phenomenon. Indeed, the intriguing longevity of semi-allogeneic fetal microchimeric cells (FMC) in parous women raises questions over their potential clinical implications. FMC have been associated with both immune-modulatory roles and participation in maternal tissue repair. Although their influence on maternal health is as yet unresolved, FMC selectively home to damaged maternal tissues and often integrate, adopting site-appropriate phenotypes. FMC features, such as plasticity and persistence in their maternal host, suggest that they likely include pluripotent, or various multipotent and committed stem and progenitor cells. Recent efforts to determine what cell types are involved have established that FMC include cells of ectodermal, endodermal, mesodermal, and perhaps trophectodermal lineages. This review details FMC phenotypes and discusses how FMC themselves may be considered a naturally occurring stem cell therapy.
Collapse
Affiliation(s)
- E Seppanen
- UQ Centre for Clinical Research, Herston Campus, University of Queensland, Brisbane, Qld. 4029, Australia
| | | | | |
Collapse
|
30
|
Lepez T, Vandewoestyne M, Deforce D. Fetal microchimeric cells in autoimmune thyroid diseases: harmful, beneficial or innocent for the thyroid gland? CHIMERISM 2013; 4:111-8. [PMID: 23723083 PMCID: PMC3921191 DOI: 10.4161/chim.25055] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Autoimmune thyroid diseases (AITD) show a female predominance, with an increased incidence in the years following parturition. Fetal microchimerism has been suggested to play a role in the pathogenesis of AITD. However, only the presence of fetal microchimeric cells in blood and in the thyroid gland of these patients has been proven, but not an actual active role in AITD. Is fetal microchimerism harmful for the thyroid gland by initiating a Graft versus Host reaction (GvHR) or being the target of a Host versus Graft reaction (HvGR)? Is fetal microchimerism beneficial for the thyroid gland by being a part of tissue repair or are fetal cells just innocent bystanders in the process of autoimmunity? This review explores every hypothesis concerning the role of fetal microchimerism in AITD.
Collapse
|
31
|
Seppanen E, Roy E, Ellis R, Bou-Gharios G, Fisk NM, Khosrotehrani K. Distant mesenchymal progenitors contribute to skin wound healing and produce collagen: evidence from a murine fetal microchimerism model. PLoS One 2013; 8:e62662. [PMID: 23650524 PMCID: PMC3641113 DOI: 10.1371/journal.pone.0062662] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 03/22/2013] [Indexed: 12/13/2022] Open
Abstract
The contribution of distant and/or bone marrow-derived endogenous mesenchymal stem cells (MSC) to skin wounds is controversial. Bone marrow transplantation experiments employed to address this have been largely confounded by radiation-resistant host-derived MSC populations. Gestationally-acquired fetal MSC are known to engraft in maternal bone marrow in all pregnancies and persist for decades. These fetal cells home to damaged maternal tissues, mirroring endogenous stem cell behavior. We used fetal microchimerism as a tool to investigate the natural homing and engraftment of distant MSC to skin wounds. Post-partum wild-type mothers that had delivered transgenic pups expressing luciferase under the collagen type I-promoter were wounded. In vivo bioluminescence imaging (BLI) was then used to track recruitment of fetal cells expressing this mesenchymal marker over 14 days of healing. Fetal cells were detected in 9/43 animals using BLI (Fisher exact p = 0.01 versus 1/43 controls). These collagen type I-expressing fetal cells were specifically recruited to maternal wounds in the initial phases of healing, peaking on day 1 (n = 43, p<0.01). This was confirmed by detection of Y-chromosome+ve fetal cells that displayed fibroblast-like morphology. Histological analyses of day 7 wounds revealed vimentin-expressing fetal cells in dermal tissue. Our results demonstrate the participation of distant mesenchymal cells in skin wounds. These data imply that endogenous MSC populations are likely recruited from bone marrow to wounds to participate in healing.
Collapse
Affiliation(s)
- Elke Seppanen
- The University of Queensland, UQ Centre for Clinical Research, Herston Campus, Brisbane, Australia
| | - Edwige Roy
- The University of Queensland, UQ Centre for Clinical Research, Herston Campus, Brisbane, Australia
| | - Rebecca Ellis
- The University of Queensland, UQ Centre for Clinical Research, Herston Campus, Brisbane, Australia
| | - George Bou-Gharios
- The University of Queensland, UQ Centre for Clinical Research, Herston Campus, Brisbane, Australia
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Nicholas M. Fisk
- The University of Queensland, UQ Centre for Clinical Research, Herston Campus, Brisbane, Australia
- Centre for Advanced Prenatal Care, Royal Brisbane and Women’s Hospital, Herston, Australia
| | - Kiarash Khosrotehrani
- The University of Queensland, UQ Centre for Clinical Research, Herston Campus, Brisbane, Australia
| |
Collapse
|
32
|
Weng SL, Chang SJ, Cheng YC, Wang HY, Wang TY, Chang MDT, Wang HW. Comparative transcriptome analysis reveals a fetal origin for mesenchymal stem cells and novel fetal surface antigens for noninvasive prenatal diagnosis. Taiwan J Obstet Gynecol 2012; 50:447-57. [PMID: 22212316 DOI: 10.1016/j.tjog.2011.10.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2011] [Indexed: 01/30/2023] Open
Abstract
OBJECTIVE Mesenchymal stem cells (MSCs) are an attractive source for providing the cells necessary for regenerating damaged tissues. Fetal MSCs (fMSCs) are known to proliferate fast and have an excellent osteogenic capacity, yet the underlying mechanisms need to be explored. A better understanding of MSCs from different anatomic origins and ages will eventually benefit cell-based therapies, as well as subsequent mechanistic studies in the field of stem cell biology. MATERIALS AND METHODS We identified the molecular signatures of fetal and adult MSCs via a meta-analytic strategy and compared the enriched canonical pathways and genetic networks within each signature. RESULTS Fetal MSCs were found to express more cell cycle genes, which is consistent with the results of wetlab functional assays. In addition, the genes involved in vasculogenesis, neurogenesis, Wnt, MAPKKK pathways, and RNA splicing were found to be enriched in fMSCs. Correlating with the overexpression of multilineage differentiation genes, fMSCs share more genes with embryonic stem cells (ESCs) and are, therefore, more primitive. Further exploration into the transcriptome similarities revealed that MSCs from umbilical cord blood (UCB) express dominant fMSC genes, but not adult genes, suggesting a fetal origin for UCB MSCs. Novel surface proteins that were dominantly expressed in fetal and UCB MSCs, but not in adult MSCs or maternal PBMCs, were also identified. CONCLUSION Our results systematically revealed the underlying genes and regulatory networks of two MSCs from unique origins, the resulting phenotypes, as well as the origin of UCB MSCs. The novel membrane proteins on the fetal MSC surface are promising candidate biomarkers for positively isolating fetal MSCs from maternal blood for noninvasive prenatal diagnosis.
Collapse
Affiliation(s)
- Shun-Long Weng
- Department of Obstetrics and Gynecology, Hsinchu Mackay Memorial Hospital, Hsinchu, Taiwan
| | | | | | | | | | | | | |
Collapse
|
33
|
Li F, Chen YZ, Miao ZN, Zheng SY, Jin J. Human placenta-derived mesenchymal stem cells with silk fibroin biomaterial in the repair of articular cartilage defects. Cell Reprogram 2012; 14:334-41. [PMID: 22816556 DOI: 10.1089/cell.2012.0002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cartilage tissue engineering requires a porous biodegradable scaffold and nonimmunogenic cells with chondrogenic potential. In this study, the ability of the placenta-derived mesenchymal stem cells (PMSCs) to grow on silk fibroin (SF) biomaterial was determined, and the potential of a SF biomaterial serving as a delivery vehicle for human PMSCs in a rabbit articular cartilage defects model was evaluated. Human PMSCs were maintained in vitro in an allogeneic mixed lymphocyte reactions (MLR) system to investigate the suppressive effects on T cell proliferation. A total of 12 healthy adult New Zealand rabbits were implanted with a PMSC/SF biomaterial complex after articular cartilage defects of the femoral condyle in the knee were established. The repair of the articular cartilage defects was observed after 4 weeks, 8 weeks, and 12 weeks. Results from the MLR indicated that human PMSCs inhibited rabbit T cell responses. Knee damage was repaired by the newly formed hyaline cartilage, and within 12 weeks there was neither degeneration nor infiltration with lymphocytes or leukocytes, and no silk fibroin biomaterial residue was detected. In conclusion, the silk fibroin biomaterial can be applied as a new scaffold for cartilage tissue engineering, and implantation of human PMSCs on the cartilage can enhance repair of articular cartilage defects in a rabbit model.
Collapse
Affiliation(s)
- Fang Li
- Department of Human Anatomy, Histology and Embryology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, P.R. China
| | | | | | | | | |
Collapse
|
34
|
Choolani M, Mahyuddin AP, Hahn S. The promise of fetal cells in maternal blood. Best Pract Res Clin Obstet Gynaecol 2012; 26:655-67. [PMID: 22795236 DOI: 10.1016/j.bpobgyn.2012.06.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 05/23/2012] [Accepted: 06/10/2012] [Indexed: 01/23/2023]
Abstract
Delaying childbirth increases the proportion of advanced maternal age pregnancies. This increases the number of pregnancies requiring invasive prenatal testing. Prenatal diagnosis of chromosomal aneuploidies and monogenic disorders requires fetal cells obtained through invasive procedures (i.e. chorionic villus sampling and amniocentesis). These procedures carry a risk of fetal loss, which causes anxiety to at-risk couples. Intact fetal cells entering maternal circulation have raised the possibility of non-invasive prenatal diagnosis. Rarity of fetal cells, however, has made it challenging. Fetal nucleated red blood cells are ideal candidate target cells because they have limited lifespan, contain true representation of fetal genotype, contain specific fetal cell identifiers (embryonic and fetal globins), and allow interrogation with chromosomal fluorescence in-situ hybridisation and possibly with array comparative genomic hybridisation. The utility of fetal nucleated red blood cells in non-invasive prenatal diagnosis has not reached clinical application because of the inconsistencies in enrichment strategies and rarity of cells.
Collapse
Affiliation(s)
- Mahesh Choolani
- Department of Obstetrics & Gynaecology, National University of Singapore, Singapore.
| | | | | |
Collapse
|
35
|
Karniychuk UU, Van Breedam W, Van Roy N, Rogel-Gaillard C, Nauwynck HJ. Demonstration of microchimerism in pregnant sows and effects of congenital PRRSV infection. Vet Res 2012; 43:19. [PMID: 22423651 PMCID: PMC3368719 DOI: 10.1186/1297-9716-43-19] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 03/16/2012] [Indexed: 11/16/2022] Open
Abstract
The presence of foreign cells within the tissue/circulation of an individual is described as microchimerism. The main purpose of the present investigation was to study if microchimerism occurs in healthy sows/fetuses and if porcine reproductive and respiratory syndrome virus (PRRSV) infection influences this phenomenon. Six dams were inoculated intranasally with PRRSV and three non-inoculated dams served as controls. Male DNA was detected in female fetal sera of all dams via PCR. Male DNA was also detected in the maternal circulation. Sex-typing FISH showed the presence of male cells in the female fetal organs and vice versa. PRRSV infection did not influence microchimerism, but might misuse maternal and sibling microchimeric cells to enter fetuses.
Collapse
Affiliation(s)
- Uladzimir U Karniychuk
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.
| | | | | | | | | |
Collapse
|
36
|
Isolation of the multipotent MSC subpopulation from human gingival fibroblasts by culturing on chitosan membranes. Biomaterials 2012; 33:2642-55. [DOI: 10.1016/j.biomaterials.2011.12.032] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 12/17/2011] [Indexed: 01/09/2023]
|
37
|
Kara RJ, Bolli P, Karakikes I, Matsunaga I, Tripodi J, Tanweer O, Altman P, Shachter NS, Nakano A, Najfeld V, Chaudhry HW. Fetal cells traffic to injured maternal myocardium and undergo cardiac differentiation. Circ Res 2011; 110:82-93. [PMID: 22082491 DOI: 10.1161/circresaha.111.249037] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
RATIONALE Fetal cells enter the maternal circulation during pregnancy and may persist in maternal tissue for decades as microchimeras. OBJECTIVE Based on clinical observations of peripartum cardiomyopathy patients and the high rate of recovery they experience from heart failure, our objective was to determine whether fetal cells can migrate to the maternal heart and differentiate to cardiac cells. METHODS AND RESULTS We report that fetal cells selectively home to injured maternal hearts and undergo differentiation into diverse cardiac lineages. Using enhanced green fluorescent protein (eGFP)-tagged fetuses, we demonstrate engraftment of multipotent fetal cells in injury zones of maternal hearts. In vivo, eGFP+ fetal cells form endothelial cells, smooth muscle cells, and cardiomyocytes. In vitro, fetal cells isolated from maternal hearts recapitulate these differentiation pathways, additionally forming vascular tubes and beating cardiomyocytes in a fusion-independent manner; ≈40% of fetal cells in the maternal heart express Caudal-related homeobox2 (Cdx2), previously associated with trophoblast stem cells, thought to solely form placenta. CONCLUSIONS Fetal maternal stem cell transfer appears to be a critical mechanism in the maternal response to cardiac injury. Furthermore, we have identified Cdx2 cells as a novel cell type for potential use in cardiovascular regenerative therapy.
Collapse
Affiliation(s)
- Rina J Kara
- Mount Sinai School of Medicine, One Gustave L Levy Place, Box 1030, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Steinkraus HB, Rothfuss H, Jones JA, Dissen E, Shefferly E, Lewis RV. The absence of detectable fetal microchimerism in nontransgenic goats (Capra aegagrus hircus) bearing transgenic offspring. J Anim Sci 2011; 90:481-8. [PMID: 21984713 DOI: 10.2527/jas.2011-4034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Regulations for the disposal of genetically engineered animals are strict due to concern for their inappropriate introduction into the food chain, and of the possible public health and environmental impacts of these organisms. Nontransgenic animals that give birth to transgenic offspring are treated as if they are transgenic due to concern of fetal cells crossing the placental barrier and residing in the mother (fetal-maternal microchimerism). Determining whether or not fetal-fetal or fetal-maternal transfer of DNA or cells occurs during caprine gestation is critical to effectively protect the public without culling animals that pose no risk. Additionally, fetal-maternal transfer, should it exist in the goat, could contraindicate the rebreeding of nontransgenic dams due to the possible transfer of fetal cells from 1 pregnancy to the fetus of subsequent pregnancies. Fetal-maternal transfer in Capra hircus has not been reported in the literature, although it has been reported in another ruminant, Bos taurus. We examined blood from nontransgenic dams that carried transgenic offspring using a PCR method sensitive enough to detect the presence of a spider silk transgene to a 1:100,000 dilution. At this sensitivity, we did not detect the occurrence of fetal-maternal transfer in 5 nontransgenic dams. Likewise, fetal-fetal transfer was not observed from a transgenic to a nontransgenic twin in utero. To test tissue-specific expression of the silk transgene, proteins purified from standard necropsy tissue from a lactating transgenic dam were examined by Western blot analysis. Silk protein expression was only observed in mammary tissue consistent with the tissue specificity of the β-casein promoter used in the transgenic construct. We report evidence collected from a limited caprine breeding pool against transfer of transgenes in utero from fetus to dam and fetus to fetus. In addition, we show evidence that the β-casein promoter in our expression construct is not expressed ectopically as previously suggested. These results suggest that transgene transfer in utero does not occur, but further study is warranted with a larger sample group to confirm these results.
Collapse
Affiliation(s)
- H B Steinkraus
- University of Wyoming, Department of Molecular Biology, Laramie 82071, USA
| | | | | | | | | | | |
Collapse
|
39
|
Boyon C, Collinet P, Boulanger L, Rubod C, Lucot JP, Vinatier D. Fetal microchimerism: benevolence or malevolence for the mother? Eur J Obstet Gynecol Reprod Biol 2011; 158:148-52. [PMID: 21664033 DOI: 10.1016/j.ejogrb.2011.05.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 05/04/2011] [Accepted: 05/10/2011] [Indexed: 01/24/2023]
Abstract
For a long time, the conventional view was that the fetus and maternal vascular system are kept separate. In fact there is a two way traffic of cells through the placenta and the transplacental passage of cells is in fact the norm. The fetal cells can persist in a wide range of woman's tissues following a pregnancy or an abortion and she becomes a chimera. Fetal cells have been found in the maternal circulation and they were shown to persist for the entire life in humans, thus demonstrating long-term engraftment and survival capabilities. Microchimerism is a subject of much interest for a number of reasons. Studies of fetal microchimerism during pregnancy may offer explanations for complications of pregnancy, such as preeclampsia, as well as insights into the pathogenesis of autoimmune diseases which usually ameliorate during pregnancy. The impact of the persistence of allogenic cells of fetal origin and of the maternal immunological response to them on the mother's health is still not clear. On the beneficial side, it has been proposed that genetically disparate fetal microchimerism provides protection against some cancers, that fetal microchimerism can afford the mother new mechanisms of protection to some diseases, that fetal microchimerism can enlarge the immunological repertoire of the mother improving her defense against aggressor. Fetal cells are often present at sites of maternal injury and may have an active role in the repair of maternal tissues.
Collapse
Affiliation(s)
- Charlotte Boyon
- Université Nord de France, Laboratoire de Spectrométrie de Masse Biologique Fondamentale et Appliquée, EA 4550, Université Lille 1, Lille, F-59650 Villeneuve d'Ascq, Service de chirurgie gynécologique, CHU Lille, F-59000 Lille, France
| | | | | | | | | | | |
Collapse
|
40
|
Huang Z, Fong CY, Gauthaman K, Sukumar P, Choolani M, Bongso A. Novel approaches to manipulating foetal cells in the maternal circulation for non-invasive prenatal diagnosis of the unborn child. J Cell Biochem 2011; 112:1475-85. [DOI: 10.1002/jcb.23084] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
41
|
Boyon C, Collinet P, Boulanger L, Vinatier D. Microchimérisme fœtal : un bien ou un mal pour le fœtus et sa mère ? ACTA ACUST UNITED AC 2011; 39:224-31. [DOI: 10.1016/j.gyobfe.2011.02.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Accepted: 12/16/2010] [Indexed: 11/25/2022]
|
42
|
Boyon C, Vinatier D. [Fetal microchimerism: self and non-self, finally who are we?]. ACTA ACUST UNITED AC 2011; 40:387-98. [PMID: 21354718 DOI: 10.1016/j.jgyn.2011.01.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2010] [Revised: 01/22/2011] [Accepted: 01/26/2011] [Indexed: 01/01/2023]
Abstract
For a long time, the conventional view was that the fetus and maternal vascular system are kept separate. In fact there is a two-way traffic of immune cells through the placenta and the transplacental passage of cells is in fact the norm. The fetal cells can persist in a wide range of woman's tissue following a pregnancy or an abortion and she becomes a chimera. Fetal cells have been found in the maternal circulation and they were shown to persist for almost three decades in humans, thus demonstrating long-term engraftment and survival capabilities. Microchimerism is a subject of much interest for a number of reasons. Studies of fetal microchimerism during pregnancy may offer explanations for complications of pregnancy, such as preeclampsia, as well as insights into the pathogenesis of autoimmune disease which usually ameliorates during pregnancy. The impact that the persistence of allogenic cells of fetal origin and the maternal immunological response to them has on the mother's health and whether it is detrimental or beneficial to the mother is still not clear. Although microchimerism has been implicated in some autoimmune diseases, fetal microchimerism is common in healthy individuals. On the beneficial side, it has been proposed that genetically disparate fetal microchimerism provides protection against some cancers, that fetal microchimerism can afford the mother new alleles of protection to some diseases she has not, that fetal microchimerism can enlarge the immunological repertoire of the mother improving her defense against aggressor. Fetal cells are often present at sites of maternal injury and may have an active role in the repair of maternal tissues.
Collapse
Affiliation(s)
- C Boyon
- FRE 3249 CNRS, cité scientifique, université Lille 1, Villeneuve d'Ascq, France
| | | |
Collapse
|
43
|
Song C, Xiang J, Tang J, Hirst DG, Zhou J, Chan KM, Li G. Thymidine kinase gene modified bone marrow mesenchymal stem cells as vehicles for antitumor therapy. Hum Gene Ther 2011; 22:439-49. [PMID: 20925460 DOI: 10.1089/hum.2010.116] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) represent an important source of cells for tissue repair. The tropism of these cells to the sites of injury and tumors has been well established. Their tumor-homing properties make BMSCs good candidates as antitumor agent delivery vehicles. In this study, we showed that BMSCs have the ability to migrate toward various cancer cells, including prostate cancer cells in vitro and in vivo and incorporating into the tumor mass. When infected with herpes simplex virus thymidine kinase (TK) gene by lentiviral transduction, TK-BMSCs maintained their tumor tropism capabilities and significantly inhibited the growth of subcutaneous PC3 prostate cancer xenografts in nude mice, in the presence of prodrug ganciclovir (GCV). Xenogenic TK-BMSCs also survived and exerted a significant antitumor effect in an animal model bearing metastastic RIF-1 (fibrosarcoma) tumor in the presence of prodrug GCV. The present study demonstrated that overexpression of TK in BMSCs did not affect their multidifferentiation potentials and their specific homing capacities toward the tumor mass, and the TK-BMSCs alone did not cause any harmful side effects in vivo. The use of TK-BMSCs as tumor-specific delivery vehicles together with controlled prodrug treatment may be a safe and novel anticancer therapy approach.
Collapse
Affiliation(s)
- Chao Song
- Stem Cells and Regeneration Program, School of Biomedical Sciences and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT Hong Kong, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
44
|
Troeger C, Perahud I, Moser S, Holzgreve W. Transplacental traffic after in utero mesenchymal stem cell transplantation. Stem Cells Dev 2011; 19:1385-92. [PMID: 20131967 DOI: 10.1089/scd.2009.0434] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Transplacental traffic of fetal progenitor and differentiated cells is a well-known phenomenon in pregnancies. We hypothesize that intrauterine stem cell transplantation leads to microchimerism in the dams and that this is gestational age-dependent. EGFP+ fetal liver-derived mesenchymal stem cell (MSC) (10(5) per fetus) were injected intraperitoneally into congeneic and allogeneic recipient fetuses at E12 versus E13.5 of murine pregnancy (56 dams). Engraftment in maternal organs was evaluated using TaqMan quantitative polymerase chain reaction (PCR) and fluorescence microscopy during pregnancy (1, 3, and 7 days after in utero transplantation [IUT]) and after delivery (1 and 4 weeks after delivery). One day after IUT donor cells were mainly found in the placenta (E12: 9/10 dams vs. E13.5: 4/8 dams) and laparotomy site (E12: 5/10 dams vs. E13.5: 4/8 dams). Three days after IUT these probabilities decreased significantly in the placenta to 3/8 and 1/3, respectively, whereas it was increased within the surgical wound to 8/8 and 2/4. One week after IUT donor cells could be detected in other single maternal organs, such as bone marrow or spleen. The surgical wound was chimeric in all dams. One week after delivery the surgical wound was still a major site of engraftment in both groups. E12 IUT resulted in detectable donor cell microchimerism in the maternal bone marrow (3/4), liver (2/4), lungs (1/4), spleen (1/4), and thymus (1/4), whereas engraftment probabilities were lower following E13.5 IUT (BM: 1/4, liver: 2/4, lungs: 1/4, spleen: 1/4, thymus: 0/4). At 4 weeks after delivery persistent microchimerism was found only after E12 IUT in various maternal organs (BM: 1/4, spleen: 1/4, lungs: 1/4) and within newly created surgical wounds (3/4), but completely not in the E13.5 group. Allogeneic IUT did also not result in any detectable long-term fetal microchimerism. An earlier IUT might lead to a higher transplacental traffic of donor MSC and persistent microchimerism within maternal tissues. Even 4 weeks after delivery, these cells are present in surgical wounds.
Collapse
Affiliation(s)
- Carolyn Troeger
- Laboratory for Prenatal Medicine, Department of Obstetrics and Gynecology, University Hospital, Basel, Switzerland.
| | | | | | | |
Collapse
|
45
|
Bou-Gharios G, Amin F, Hill P, Nakamura H, Maxwell P, Fisk NM. Microchimeric Fetal Cells Are Recruited to Maternal Kidney following Injury and Activate Collagen Type I Transcription. Cells Tissues Organs 2011; 193:379-92. [DOI: 10.1159/000321172] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2010] [Indexed: 01/15/2023] Open
|
46
|
Witherick J, Wilkins A, Scolding N, Kemp K. Mechanisms of oxidative damage in multiple sclerosis and a cell therapy approach to treatment. Autoimmune Dis 2010; 2011:164608. [PMID: 21197107 PMCID: PMC3010615 DOI: 10.4061/2011/164608] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 11/10/2010] [Indexed: 12/14/2022] Open
Abstract
Although significant advances have recently been made in the understanding and treatment of multiple sclerosis, reduction of long-term disability remains a key goal. Evidence suggests that inflammation and oxidative stress within the central nervous system are major causes of ongoing tissue damage in the disease. Invading inflammatory cells, as well as resident central nervous system cells, release a number of reactive oxygen and nitrogen species which cause demyelination and axonal destruction, the pathological hallmarks of multiple sclerosis. Reduction in oxidative damage is an important therapeutic strategy to slow or halt disease processes. Many drugs in clinical practice or currently in trial target this mechanism. Cell-based therapies offer an alternative source of antioxidant capability. Classically thought of as being important for myelin or cell replacement in multiple sclerosis, stem cells may, however, have a more important role as providers of supporting factors or direct attenuators of the disease. In this paper we focus on the antioxidant properties of mesenchymal stem cells and discuss their potential importance as a cell-based therapy for multiple sclerosis.
Collapse
Affiliation(s)
- Jonathan Witherick
- Multiple Sclerosis and Stem Cell Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol BS16 1LE, UK
| | - Alastair Wilkins
- Multiple Sclerosis and Stem Cell Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol BS16 1LE, UK
| | - Neil Scolding
- Multiple Sclerosis and Stem Cell Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol BS16 1LE, UK
| | - Kevin Kemp
- Multiple Sclerosis and Stem Cell Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol BS16 1LE, UK
| |
Collapse
|
47
|
Lee ESM, Bou-Gharios G, Seppanen E, Khosrotehrani K, Fisk NM. Fetal stem cell microchimerism: natural-born healers or killers? Mol Hum Reprod 2010; 16:869-78. [PMID: 20663958 DOI: 10.1093/molehr/gaq067] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Eddy S M Lee
- University of Queensland Centre for Clinical Research, University of Queensland, Herston campus, Brisbane 4029, Australia
| | | | | | | | | |
Collapse
|
48
|
Hu KX, Sun QY, Guo M, Ai HS. The radiation protection and therapy effects of mesenchymal stem cells in mice with acute radiation injury. Br J Radiol 2010; 83:52-8. [PMID: 20139249 DOI: 10.1259/bjr/61042310] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The aim of this study was to investigate the effects and mechanisms of mesenchymal stem cells (MSCs) on haematopoietic reconstitution in reducing bone marrow cell apoptosis effects in irradiated mice, and to research the safe and effective dosage of MSCs in mice with total body irradiation (TBI). After BALB/c mice were irradiated with 5.5 Gy cobalt-60 gamma-rays, the following were observed: peripheral blood cell count, apoptosis rate, cell cycle, colony-forming unit-granulocyte macrophage (CFU-GM) and colony-forming unit-fibroblast (CFU-F) counts of bone marrow cells and pathological changes in the medulla. The survival of mice infused with three doses of MSCs after 8.0 Gy or 10 Gy TBI was examined. The blood cells recovered rapidly in the MSC groups. The apoptotic ratio of bone marrow cells in the control group was higher at 24 h after radiation. A lower ratio of G0/G1 cell cycle phases and a higher ratio of G2/M and S phases, as well as a greater number of haematopoietic islands and megalokaryocytes in the bone marrow, were observed in the MSC-treated groups. MSCs induced recovery of CFU-GM and CFU-GM and improved the survival of mice after 8 Gy TBI, but 1.5 x 10(8) kg(-1) of MSCs increased mortality. These results indicate that MSCs protected and treated irradiated mice by inducing haematopoiesis and reducing apoptosis. MSCs may be a succedaneous or intensive method of haematopoietic stem cell transplantation under certain radiation dosages, and could provide a valuable strategy for acute radiation syndrome.
Collapse
Affiliation(s)
- K X Hu
- Department of Haematology and Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | | | | | | |
Collapse
|
49
|
Kemp K, Mallam E, Scolding N, Wilkins A. Stem cells in genetic myelin disorders. Regen Med 2010; 5:425-39. [DOI: 10.2217/rme.10.10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The genetic myelin disorders are a range of diseases that manifest with severe neurological problems, often from infancy. It has been postulated for some time that stem cells might be an effective treatment for these disorders, primarily as agents to restore dysfunctional or lost myelin. Stem cells, however, may offer a wider range of therapeutic potential, for instance as vehicles to replace abnormal enzymes or genes, or to provide trophic support for residual CNS tissue. This article will review several of the more common genetic myelin disorders and currently available therapies, including bone marrow transplantation for adrenoleukodystrophy. Specific stem cell subtypes and their relevance to potential therapeutic use will be discussed and stem cell transplantation in animal model studies will also be reviewed.
Collapse
Affiliation(s)
- Kevin Kemp
- MS & Stem Cell Laboratories, Burden Centre, Frenchay Hospital, Bristol, UK
- Department of Neurology, Frenchay Hospital, Bristol, UK
| | - Elizabeth Mallam
- MS & Stem Cell Laboratories, Burden Centre, Frenchay Hospital, Bristol, UK
- Department of Neurology, Frenchay Hospital, Bristol, UK
| | - Neil Scolding
- MS & Stem Cell Laboratories, Burden Centre, Frenchay Hospital, Bristol, UK
- Department of Neurology, Frenchay Hospital, Bristol, UK
| | | |
Collapse
|
50
|
Gotherstrom C, Chan J, O'Donoghue K, Fisk NM. Identification of candidate surface antigens for non-invasive prenatal diagnosis by comparative global gene expression on human fetal mesenchymal stem cells. Mol Hum Reprod 2010; 16:472-80. [DOI: 10.1093/molehr/gaq020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|