141 results on '"Schoeberlein A"'
Search Results
2. Engraftment kinetics of human cord blood and murine fetal liver stem cells following in utero transplantation into immunodeficient mice.
- Author
-
Schoeberlein A, Schatt S, Troeger C, Surbek D, Holzgreve W, and Hahn S
- Subjects
- Animals, Antigens, CD34 biosynthesis, Cell Lineage, Cell Separation, Cell Transplantation, Chimerism, Flow Cytometry, Globins metabolism, Hematopoietic Stem Cell Transplantation methods, Hematopoietic Stem Cells cytology, Humans, Kinetics, Liver Extracts, Mice, Mice, Inbred C57BL, Mice, SCID, Time Factors, Fetal Blood cytology, Liver embryology, Stem Cells cytology
- Abstract
This study was undertaken to evaluate the kinetics of engraftment after in utero transplantation of murine fetal liver and human cord blood stem cells in the nonobese diabetic/severe combined immunodeficient (NOD/SCID) mouse model. NOD/SCID fetuses were injected with murine fetal liver or human cord blood CD34+ cells at day 13.5 of gestation. Frequencies of donor cells were analyzed by flow cytometry up to 48 h post transplantation and 4-16 weeks postnatally. Hematopoietic multilineage reconstitution capacity was assessed. Both types of donor cells home rapidly. However, the frequency of human cord blood stem cells rapidly diminished while the murine fetal liver stem cells expanded over time, resulting in multilineage hematopoietic reconstitution. Differences in long-term reconstitution of allogeneic versus xenogeneic donor cells were ascribed to the inability of the human cells to self-renew and differentiate in the fetal mouse environment, demonstrating the limitations of this commonly used xenograph.
- Published
- 2004
- Full Text
- View/download PDF
3. In vitro-microenvironment directs preconditioning of human chorion derived MSC promoting differentiation of OPC-like cells.
- Author
-
Periasamy, Ramesh, Surbek, Daniel V., and Schoeberlein, Andreina
- Subjects
MESENCHYMAL stem cell differentiation ,OLIGODENDROGLIA ,PROGENITOR cells ,CHORION ,BRAIN injuries ,PROTEIN expression - Abstract
The loss of oligodendrocyte progenitor cells (OPC) is a hallmark of perinatal brain injury. Our aim was to develop an in vitro culture condition for human chorion-derived mesenchymal stem cells (MSC) that enhances their stem cell properties and their capability to differentiate towards OPC-like cells. MSC were grown either in serum replacement medium (SRM) or serum-containing medium (SM) and tested for their morphology, proliferation, secretome, migration, protein expression and differentiation into OPC-like cells. MSC cultured in SRM condition have distinct morphology/protein expression profile, increased cell proliferation/migration and capacity to differentiate into OPC-like cells. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
4. Engraftment kinetics of human cord blood and murine fetal liver stem cells following in utero transplantation into immunodeficient mice
- Author
-
Sinuhe Hahn, Carolyn Troeger, Daniel Surbek, Stephan Schatt, Wolfgang Holzgreve, and Andreina Schoeberlein
- Subjects
Time Factors ,Cell Transplantation ,CD34 ,Liver Stem Cell ,Antigens, CD34 ,Nod ,Cell Separation ,Mice, SCID ,Biology ,Chimerism ,In utero transplantation ,Andrology ,Mice ,Animals ,Humans ,Cell Lineage ,Fetus ,Stem Cells ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Hematology ,Fetal Blood ,Flow Cytometry ,Hematopoietic Stem Cells ,Globins ,Mice, Inbred C57BL ,Haematopoiesis ,Kinetics ,Liver ,Cord blood ,Immunology ,Stem cell ,Liver Extracts ,Developmental Biology - Abstract
This study was undertaken to evaluate the kinetics of engraftment after in utero transplantation of murine fetal liver and human cord blood stem cells in the nonobese diabetic/severe combined immunodeficient (NOD/SCID) mouse model. NOD/SCID fetuses were injected with murine fetal liver or human cord blood CD34 � cells at day 13.5 of gestation. Frequencies of donor cells were analyzed by flow cytometry up to 48 h post transplantation and 4–16 weeks postnatally. Hematopoietic multilineage reconstitution capacity was assessed. Both types of donor cells home rapidly. However, the frequency of human cord blood stem cells rapidly diminished while the murine fetal liver stem cells expanded over time, resulting in multilineage hematopoietic reconstitution. Differences in long-term reconstitution of allogeneic versus xenogeneic donor cells were ascribed to the inability of the human cells to self-renew and differentiate in the fetal mouse environment, demonstrating the limitations of this commonly used xenograph.
- Published
- 2005
5. Fetal gene therapy: Opportunities and risks
- Author
-
Wagner, Anna M., Schoeberlein, Andreina, and Surbek, Daniel
- Subjects
- *
PREGNANCY complications , *GENETIC disorder diagnosis , *GENE therapy , *HEMATOPOIETIC stem cells , *STEM cell transplantation , *MOLECULAR genetics - Abstract
Abstract: Advances in human prenatal medicine and molecular genetics have allowed the diagnosis of many genetic diseases early in gestation. In-utero transplantation of allogeneic hematopoietic stem cells (HSC) has been successfully used as a therapy in different animal models and recently also in human fetuses. Unfortunately, clinical success of this novel treatment is limited by the lack of donor cell engraftment in non-immunocompromised hosts and is thus restricted to diseases where the fetus is affected by severe immunodeficiency. Gene therapy using genetically modified autologous HSC circumvents allogeneic HLA barriers and constitutes one of the most promising new approaches to correct genetic deficits in the fetus. Recent developments of strategies to overcome failure of efficient transduction of quiescent hematopoietic cells include the use of new vector constructs and transduction protocols. These improvements open new perspectives for gene therapy in general and for prenatal gene transfer in particular. The fetus may be especially susceptible for successful gene therapy due to the immunologic naiveté of the immature hematopoietic system during gestation, precluding an immune reaction towards the transgene. Ethical issues, in particular those regarding treatment safety, must be taken into account before clinical trials with fetal gene therapy in human pregnancies can be initiated. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
6. Perinatal stem-cell and gene therapy for hemoglobinopathies.
- Author
-
Surbek, Daniel, Schoeberlein, Andreina, and Wagner, Anna
- Abstract
Summary: Most genetic diseases of the lymphohematopoietic system, including hemoglobinopathies, can now be diagnosed early in gestation. However, as yet, prenatal treatment is not available. Postnatal therapy by hematopoietic stem cell (HSC) transplantation from bone marrow, mobilized peripheral blood, or umbilical cord blood is possible for several of these diseases, in particular for the hemoglobinopathies, but is often limited by a lack of histocompatible donors, severe treatment-associated morbidity, and preexisting organ damage that developed before birth. In-utero transplantation of allogeneic HSC has been performed successfully in various animal models and recently in humans. However, the clinical success of this novel treatment is limited to diseases in which the fetus is affected by severe immunodeficiency. The lack of donor cell engraftment in nonimmunocompromised hosts is thought to be due to immunologic barriers, as well as to competitive fetal marrow population by host HSCs. Among the possible strategies to circumvent allogeneic HLA barriers, the use of gene therapy by genetically corrected autologous HSCs in the fetus is one of the most promising approaches. The recent development of strategies to overcome failure of efficient transduction of quiescent hematopoietic cells using new vector constructs and transduction protocols opens new perspectives for gene therapy in general, as well as for prenatal gene transfer in particular. The fetus might be especially susceptible for successful gene therapy approaches because of the developing, expanding hematopoietic system during gestation and the immunologic naiveté early in gestation, precluding immune reaction towards the transgene by inducing tolerance. Ethical issues, in particular regarding treatment safety, must be addressed more closely before clinical trials with fetal gene therapy in human pregnancies can be initiated. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
7. Placental mesenchymal stem cells as potential autologous graft for pre- and perinatal neuroregeneration.
- Author
-
Portmann-Lanz, C. Bettina, Schoeberlein, Andreina, Huber, Alexander, Sager, Ruth, Malek, Antoine, Holzgreve, Wolfgang, and Surbek, Daniel V.
- Subjects
STEM cells ,CELL differentiation ,FETAL membranes ,TISSUES ,PLACENTA ,FLUORESCENCE ,GESTATIONAL age ,CELL culture ,CELLS - Abstract
Objective: Mesenchymal stem cells (MSCs) have a broad differentiation potential. We aimed to determine if MSCs are present in fetal membranes and placental tissue and to assess their potential to differentiate into neurogenic and mesodermal lineages. Study design: MSCs isolated from first and third trimester chorion and amnion and first trimester chorionic villi and characterized morphologically and by flourescence-activated cell sorting analysis. Their ability to mature under different culture conditions into various cells of mesodermal and neuroectodermal cell lines was assessed by immuno- and cytochemical staining. Results: Independent of gestational age, cells isolated from fetal membranes and placenta showed typical MSC phenotype (positive for CD166, CD105, CD90, CD73, CD49e, CD44, CD29, CD 13, MHC I; negative for CD 14, CD34, CD45, MHC II) and were able to differentiate into mesodermal cells expressing cell markers/cytologic staining consistent with mature chondroblasts, osteoblasts, adipocytes, or myocytes and into neuronal cells presenting markers of various stages of maturation. The differentiation pattern was mainly dependent on cell type. Conclusion: Mesenchymal cells from chorion, amnion, and villous stroma can be differentiated into neurogenic, chondrogenic, osteogenic, adipogenic, and myogenic lineage. Placental tissue obtained during prenatal chorionic villous sampling or at delivery might be an ideal source for autologous stem cell graft for peripartum neuroregeneration and other clinical issues. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
8. Tissue-specific engraftment after in utero transplantation of allogeneic mesenchymal stem cells into sheep fetuses.
- Author
-
Schoeberlein, Andreina, Holzgreve, Wolfgang, Dudler, Lisbeth, Hahn, Sinuhe, and Surbek, Daniel V.
- Subjects
TRANSPLANTATION of organs, tissues, etc. ,STEM cells ,GENETIC disorders ,DISEASES ,PRENATAL diagnosis ,OBSTETRICAL diagnosis ,SHEEP as laboratory animals - Abstract
Objective: Mesenchymal stem cells (MSC) have multiorgan differentiation capacity, providing the potential for prenatal treatment of genetic disorders. We address the question if in utero transplantation of MSC results in short-term organ-specific engraftment in the fetal sheep. Study design: Sheep fetal liver-derived MSC selected by adherence culture (passage 1) were transplantated into the fetal peritoneal cavity with ultrasound-guidance (mean gestational age, 59 days). After 14 days recipient fetuses were analyzed by fluorescence-activated cell sorting (FACS), real-time polymerase chain reaction (PCR), and immunohistochemistry. Results: Fetuses (n = 11) were transplanted with 7.7×10
6 MSCs (mean). All surviving fetuses (n = 5) showed engraftment with mean levels of 3.2% (lung), 0.8% (spleen), 0.6% (liver, brain), 0.4% (bone marrow), 0.1% (blood, thymus), and <0.1% (kidneys) by flow cytometry. Immunohistochemistry showed organ-specific distribution. Conclusion: In utero transplantation of allogeneic MSC results in low level, multiorgan engraftment at 14 days post transplant. This supports the potential of in utero MSC transplantation for the treatment of nonhematopoietic genetic disorders of the fetus. [ABSTRACT FROM AUTHOR]- Published
- 2005
- Full Text
- View/download PDF
9. In utero transplantation of autologous and allogeneic fetal liver stem cells in ovine fetuses.
- Author
-
Schoeberlein, Andreina, Holzgreve, Wolfgang, Dudler, Lisbeth, Hahn, Sinuhe, and Surbek, Daniel V.
- Subjects
FETAL liver cells ,STEM cells ,STEM cell transplantation ,TRANSPLANTATION of organs, tissues, etc. ,AUTOTRANSPLANTATION ,SHEEP as laboratory animals ,ANIMAL models in research ,MAJOR histocompatibility complex - Abstract
Objective: The purpose of this study was to assess the feasibility of autologous stem cell transplantation in fetal sheep and to compare short-term engraftment of allogeneic and autologous fetal liver stem cells in an immunocompetent large animal model. Study design: Fetal liver stem cells were collected from preimmune sheep fetuses with an open or ultrasound-guided technique. After being labeled with PKH26, the cells were transplanted intraperitoneally into allogeneic and autologous fetal recipients at 48 to 64 days of gestation. Engraftment was determined by flow cytometry and real-time polymerase chain reaction 1 to 2 weeks after transplantation. Results: Fetal loss rate was 29% (allogeneic transplantation) and 73% (autologous transplantation). Engraftment of donor cells was found in all fetuses, with a level of ≤4.7% in fetal liver, spleen, bone marrow, blood and thymus. Overall, there was no difference between allogeneic and autologous grafts. Conclusion: Autologous in utero transplantation of fetal liver stem cells in fetal sheep is feasible, but yields a high loss rate. Differences in the major histocompatibility complex between donor and recipient seems not to have a major impact on stem cell engraftment early in gestation; major histocompatibility complex—independent donor/host competition might be responsible for low engraftment in immunocompetent recipients. [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
10. Ultrasound-guided stem cell sampling from the early ovine fetus for prenatal ex vivo gene therapy.
- Author
-
Surbek, Daniel V., Young, Alan, Danzer, Enrico, Schoeberlein, Andreina, Dudler, Lisbeth, and Holzgreve, Wolfgang
- Subjects
PREGNANCY ,GENE therapy ,STEM cells ,ERYTHROCYTES ,ANIMAL experimentation ,COLLECTION & preservation of biological specimens ,COMPARATIVE studies ,FETUS ,FETAL ultrasonic imaging ,HEMATOPOIETIC stem cells ,LIVER ,RESEARCH methodology ,MEDICAL cooperation ,FIRST trimester of pregnancy ,PRENATAL care ,RESEARCH ,SHEEP ,PILOT projects ,CYTOMETRY ,EVALUATION research ,MEDICAL suction ,COMPUTER-assisted surgery - Abstract
Objective: Prenatal ex vivo gene therapy might be an effective and safe strategy with which to treat severe genetic disorders in utero. For this purpose, autologous fetal stem cells must be collected before the second trimester, transfected in vitro, and transplanted back to the fetus. The aim of this study was to determine whether stem cells can be sampled from the first trimester fetal liver in ongoing gestation.Study Design: Fetal liver stem cell sampling was performed in 21 ovine fetuses. Pregnant ewes at 57 +/- 2 gestational days were generally anesthesized. A 20-gauge needle was inserted transcutaneously into the fetal liver under ultrasound guidance. Fetal liver cells were sampled by suction. The numbers of nucleated cells and progenitor/stem cells were determined.Results: All 21 fetuses showed normal heart rate 5 minutes after the procedure. A mean (+/-SEM) of 2.07 +/- 0.5 x 10(7) nucleated cells and 172 +/- 53 colony-forming units per 10(5) cells (hematopoietic progenitors/stem cells) were collected. Fetal loss rate at term was 7 of 21 fetuses (33%).Conclusion: This study shows that fetal liver cells can be collected in the early fetus with an ultrasound-guided technique. The number of fetal liver cells that are collectable is large enough for autologous transplantation and engraftment of genetically engineered (transfected) stem cells. [ABSTRACT FROM AUTHOR]- Published
- 2002
- Full Text
- View/download PDF
11. Human Umbilical Cord-Mesenchymal Stem Cells Promote Extracellular Matrix Remodeling in Microglia.
- Author
-
Lombardo, Marta Tiffany, Gabrielli, Martina, Julien-Marsollier, Florence, Faivre, Valérie, Le Charpentier, Tifenn, Bokobza, Cindy, D'Aliberti, Deborah, Pelizzi, Nicola, Halimi, Camilla, Spinelli, Silvia, Van Steenwinckel, Juliette, Verderio, Elisabetta A. M., Gressens, Pierre, Piazza, Rocco, and Verderio, Claudia
- Subjects
MESENCHYMAL stem cells ,HUMAN stem cells ,EXTRACELLULAR matrix ,STEM cells ,CELL migration - Abstract
Human mesenchymal stem cells modulate the immune response and are good candidates for cell therapy in neuroinflammatory brain disorders affecting both adult and premature infants. Recent evidence indicates that through their secretome, mesenchymal stem cells direct microglia, brain-resident immune cells, toward pro-regenerative functions, but the mechanisms underlying microglial phenotypic transition are still under investigation. Using an in vitro coculture approach combined with transcriptomic analysis, we identified the extracellular matrix as the most relevant pathway altered by the human mesenchymal stem cell secretome in the response of microglia to inflammatory cytokines. We confirmed extracellular matrix remodeling in microglia exposed to the mesenchymal stem cell secretome via immunofluorescence analysis of the matrix component fibronectin and the extracellular crosslinking enzyme transglutaminase-2. Furthermore, an analysis of hallmark microglial functions revealed that changes in the extracellular matrix enhance ruffle formation by microglia and cell motility. These findings point to extracellular matrix changes, associated plasma membrane remodeling, and enhanced microglial migration as novel mechanisms by which mesenchymal stem cells contribute to the pro-regenerative microglial transition. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Intranasally Administered Exosomes from Umbilical Cord Stem Cells Have Preventive Neuroprotective Effects and Contribute to Functional Recovery after Perinatal Brain Injury.
- Author
-
Thomi, Gierin, Joerger-Messerli, Marianne, Haesler, Valérie, Muri, Lukas, Surbek, Daniel, and Schoeberlein, Andreina
- Subjects
EXOSOMES ,BRAIN injuries ,UMBILICAL cord ,STEM cells ,LEARNING in animals ,CELL death - Abstract
Perinatal brain injury (PBI) in preterm birth is associated with substantial injury and dysmaturation of white and gray matter, and can lead to severe neurodevelopmental deficits. Mesenchymal stromal cells (MSC) have been suggested to have neuroprotective effects in perinatal brain injury, in part through the release of extracellular vesicles like exosomes. We aimed to evaluate the neuroprotective effects of intranasally administered MSC-derived exosomes and their potential to improve neurodevelopmental outcome after PBI. Exosomes were isolated from human Wharton's jelly MSC supernatant using ultracentrifugation. Two days old Wistar rat pups were subjected to PBI by a combination of inflammation and hypoxia-ischemia. Exosomes were intranasally administered after the induction of inflammation and prior to ischemia, which was followed by hypoxia. Infrared-labeled exosomes were intranasally administered to track their distribution with a LI-COR scanner. Acute oligodendrocyte- and neuron-specific cell death was analyzed 24 h after injury in animals with or without MSC exosome application using terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay and immunohistochemical counterstaining. Myelination, mature oligodendroglial and neuronal cell counts were assessed on postnatal day 11 using immunohistochemistry, Western blot or RT-PCR. Morris water maze assay was used to evaluate the effect of MSC exosomes on long-term neurodevelopmental outcome 4 weeks after injury. We found that intranasally administered exosomes reached the frontal part of the brain within 30 min after administration and distributed throughout the whole brain after 3 h. While PBI was not associated with oligodendrocyte-specific cell death, it induced significant neuron-specific cell death which was substantially reduced upon MSC exosome application prior to ischemia. MSC exosomes rescued normal myelination, mature oligodendroglial and neuronal cell counts which were impaired after PBI. Finally, the application of MSC exosomes significantly improved learning ability in animals with PBI. In conclusion, MSC exosomes represent a novel prevention strategy with substantial clinical potential as they can be administered intranasally, prevent gray and white matter alterations and improve long-term neurodevelopmental outcome after PBI. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
13. A Cellular Model of Amyotrophic Lateral Sclerosis to Study the Therapeutic Effects of Extracellular Vesicles from Adipose Mesenchymal Stem Cells on Microglial Activation.
- Author
-
Dabrowska, Sylwia, Turano, Ermanna, Scambi, Ilaria, Virla, Federica, Nodari, Alice, Pezzini, Francesco, Galiè, Mirco, Bonetti, Bruno, and Mariotti, Raffaella
- Subjects
AMYOTROPHIC lateral sclerosis ,MICROGLIA ,MESENCHYMAL stem cells ,EXTRACELLULAR vesicles ,TREATMENT effectiveness ,CELL death - Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive degeneration of upper and lower motor neurons (MNs) in the brain and spinal cord, leading to progressive paralysis and death. Increasing evidence indicates that neuroinflammation plays an important role in ALS's pathogenesis and disease progression. Neuroinflammatory responses, primarily driven by activated microglia and astrocytes, and followed by infiltrating peripheral immune cells, contribute to exacerbate/accelerate MN death. In particular, the role of the microglia in ALS remains unclear, partly due to the lack of experimental models that can fully recapitulate the complexity of ALS's pathology. In this study, we developed and characterized a microglial cell line, SIM-A9-expressing human mutant protein Cu
+ /Zn+ superoxide dismutase_1 (SIM-A9hSOD1(G93A)), as a suitable model in vitro mimicking the microglia activity in ALS. The expression of hSOD1(G93A) in SIM-A9 cells induced a change in their metabolic activity, causing polarization into a pro-inflammatory phenotype and enhancing reactive oxygen species production, which is known to activate cell death processes and apoptosis. Afterward, we used our microglial model as an experimental set-up to investigate the therapeutic action of extracellular vesicles isolated from adipose mesenchymal stem cells (ASC-EVs). ASC-EVs represent a promising therapeutic treatment for ALS due to their neuroprotective and immunomodulatory properties. Here, we demonstrated that treatment with ASC-EVs is able to modulate activated ALS microglia, reducing their metabolic activity and polarizing their phenotype toward an anti-inflammatory one through a mechanism of reduction of reactive oxygen species. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
14. Human adipose tissue-derived stem cell extracellular vesicles attenuate ocular hypertension-induced retinal ganglion cell damage by inhibiting microglia- TLR4/MAPK/NF-κB proinflammatory cascade signaling.
- Author
-
Ji, Shangli, Peng, Yanfang, Liu, Jian, Xu, Pang, and Tang, Shibo
- Subjects
EXTRACELLULAR vesicles ,RETINAL ganglion cells ,ADIPOSE tissues ,STEM cells ,OPTIC nerve injuries ,GLAUCOMA ,MESENCHYMAL stem cells - Abstract
Microglia-mediated neuroinflammatory responses are recognized as a predominant factor during high intraocular pressure (IOP)-induced retinal and optic nerve injury along with potential therapeutic targets for the disease. Our previous research indicated that mesenchymal stem cell (MSC) treatment could reduce high IOP-induced neuroinflammatory responses through the TLR4 pathway in a rat model without apparent cell replacement and differentiation, suggesting that the anti-neuroinflammatory properties of MSCs are potentially mediated by paracrine signaling. This study aimed to evaluate the anti-neuroinflammatory effect of human adipose tissue-derived extracellular vesicles (ADSC-EVs) in microbead-induced ocular hypertension (OHT) animals and to explore the underlying mechanism since extracellular vesicles (EVs) are the primary transporters for cell secretory action. The anti-neuroinflammatory effect of ADSC-EVs on LPS-stimulated BV-2 cells in vitro and OHT-induced retinal and optic nerve injury in vivo was investigated. According to the in vitro research, ADSC-EV treatment reduced LPS-induced microglial activation and the TLR4/NF-κB proinflammatory cascade response axis in BV-2 cells, such as CD68, iNOS, TNF-α, IL-6, and IL-1β, TLR4, p-38 MAPK, NF-κB. According to the in vivo data, intravitreal injection of ADSC-EVs promoted RGC survival and function, reduced microglial activation, microglial-derived neuroinflammatory responses, and TLR4/MAPK/NF-κB proinflammatory cascade response axis in the OHT mice. Our findings provide preliminary evidence for the RGC protective and microglia-associated neuroinflammatory reduction effects of ADSC-EVs by inhibiting the TLR4/MAPK/NF-κB proinflammatory cascade response in OHT mice, indicating the therapeutic potential ADSC-EVs or adjunctive therapy for glaucoma. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. Allogeneic Mesenchymal Stem Cells After In Vivo Transplantation: A Review.
- Author
-
Asserson, Derek B.
- Subjects
ORGANS (Anatomy) ,STEM cells ,MESENCHYMAL stem cells - Abstract
Autologous mesenchymal stem cells (MSCs) are ideal for tissue regeneration because of their ability to circumvent host rejection, but their procurement and processing present logistical and time-sensitive challenges. Allogeneic MSCs provide an alternative cell-based therapy capable of positively affecting all human organ systems, and can be readily available. Extensive research has been conducted in the treatment of autoimmune, degenerative, and inflammatory diseases with such stem cells, and has demonstrated predominantly safe outcomes with minimal complications. Nevertheless, continued clinical trials are necessary to ascertain optimal harvest and transplant techniques. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
16. The role of stem cells in obstetrics and gynecology: A systematic review.
- Author
-
Mirani, Putri, Legiran, Kesty, Cindy, and Andrina, Hana
- Subjects
STEM cell transplantation ,MATERNAL health services ,ONLINE information services ,ENDOMETRIOSIS ,LEIOMYOSARCOMA ,ENDOMETRIAL diseases ,X-linked genetic disorders ,SYSTEMATIC reviews ,UTERINE fibroids ,QUALITATIVE research ,URINARY stress incontinence ,CHROMOSOME abnormalities ,GYNECOLOGIC care ,MEDLINE ,ANAL sex ,WOMEN'S health ,ENDOMETRIUM ,EPIGENOMICS - Published
- 2023
- Full Text
- View/download PDF
17. Secretome as a Tool to Treat Neurological Conditions: Are We Ready?
- Author
-
da Silva, Andreia Valente, Serrenho, Inês, Araújo, Beatriz, Carvalho, Alexandre Martins, and Baltazar, Graça
- Subjects
NEUROLOGICAL disorders ,MESENCHYMAL stem cells ,ENCEPHALITIS ,REGENERATIVE medicine ,STEM cells - Abstract
Due to their characteristics, mesenchymal stem cells (MSCs) are considered a potential therapy for brain tissue injury or degeneration. Nevertheless, despite the promising results observed, there has been a growing interest in the use of cell-free therapies in regenerative medicine, such as the use of stem cell secretome. This review provides an in-depth compilation of data regarding the secretome composition, protocols used for its preparation, as well as existing information on the impact of secretome administration on various brain conditions, pointing out gaps and highlighting relevant findings. Moreover, due to the ability of MSCs to respond differently depending on their microenvironment, preconditioning of MSCs has been used to modulate their composition and, consequently, their therapeutic potential. The different strategies used to modulate the MSC secretome were also reviewed. Although secretome administration was effective in improving functional impairments, regeneration, neuroprotection, and reducing inflammation in brain tissue, a high variability in secretome preparation and administration was identified, compromising the transposition of preclinical data to clinical studies. Indeed, there are no reports of the use of secretome in clinical trials. Despite the existing limitations and lack of clinical data, secretome administration is a potential tool for the treatment of various diseases that impact the CNS. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
18. Amniotic Fluid and Placental Membranes as Sources of Stem Cells: Progress and Challenges 2.0.
- Author
-
Maraldi, Tullia and Russo, Valentina
- Subjects
STEM cells ,AMNIOTIC liquid ,PLACENTA ,IMMUNE response - Abstract
This document is a summary of a special issue of the International Journal of Molecular Sciences that focuses on the therapeutic potential of perinatal stem cells, specifically amniotic fluid and placental membranes, in various pathological conditions. The article highlights the advantages of using amniotic cells, such as their lack of tumorigenicity and ethical concerns, as well as their ability to modulate immune responses. It also discusses the use of three-dimensional culturing techniques to enhance the properties of amniotic cells and the potential of amniotic cells in treating inflammation-related diseases. The document concludes by acknowledging the challenges and limitations associated with using amniotic stem cells and the need for further research in this field. [Extracted from the article]
- Published
- 2023
- Full Text
- View/download PDF
19. Multipotent fetal stem cells in reproductive biology research.
- Author
-
Rosner, Margit, Horer, Stefanie, Feichtinger, Michael, and Hengstschläger, Markus
- Subjects
MULTIPOTENT stem cells ,GERM cells ,CYTOLOGY ,PLURIPOTENT stem cells ,STEM cells ,AMNIOTIC liquid - Abstract
Due to the limited accessibility of the in vivo situation, the scarcity of the human tissue, legal constraints, and ethical considerations, the underlying molecular mechanisms of disorders, such as preeclampsia, the pathological consequences of fetomaternal microchimerism, or infertility, are still not fully understood. And although substantial progress has already been made, the therapeutic strategies for reproductive system diseases are still facing limitations. In the recent years, it became more and more evident that stem cells are powerful tools for basic research in human reproduction and stem cell-based approaches moved into the center of endeavors to establish new clinical concepts. Multipotent fetal stem cells derived from the amniotic fluid, amniotic membrane, chorion leave, Wharton´s jelly, or placenta came to the fore because they are easy to acquire, are not associated with ethical concerns or covered by strict legal restrictions, and can be banked for autologous utilization later in life. Compared to adult stem cells, they exhibit a significantly higher differentiation potential and are much easier to propagate in vitro. Compared to pluripotent stem cells, they harbor less mutations, are not tumorigenic, and exhibit low immunogenicity. Studies on multipotent fetal stem cells can be invaluable to gain knowledge on the development of dysfunctional fetal cell types, to characterize the fetal stem cells migrating into the body of a pregnant woman in the context of fetomaternal microchimerism, and to obtain a more comprehensive picture of germ cell development in the course of in vitro differentiation experiments. The in vivo transplantation of fetal stem cells or their paracrine factors can mediate therapeutic effects in preeclampsia and can restore reproductive organ functions. Together with the use of fetal stem cell-derived gametes, such strategies could once help individuals, who do not develop functional gametes, to conceive genetically related children. Although there is still a long way to go, these developments regarding the usage of multipotent fetal stem cells in the clinic should continuously be accompanied by a wide and detailed ethical discussion. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
20. Small Extracellular Vesicles' miRNAs: Biomarkers and Therapeutics for Neurodegenerative Diseases.
- Author
-
Lim, Wei Qing, Michelle Luk, Kie Hoon, Lee, Kah Yee, Nurul, Nasuha, Loh, Sin Jade, Yeow, Zhen Xiong, Wong, Qi Xuan, Daniel Looi, Qi Hao, Chong, Pan Pan, How, Chee Wun, Hamzah, Sharina, and Foo, Jhi Biau
- Subjects
EXTRACELLULAR vesicles ,NEURODEGENERATION ,MICRORNA ,DISEASE progression ,STEM cells - Abstract
Neurodegenerative diseases are critical in the healthcare system as patients suffer from progressive diseases despite currently available drug management. Indeed, the growing ageing population will burden the country's healthcare system and the caretakers. Thus, there is a need for new management that could stop or reverse the progression of neurodegenerative diseases. Stem cells possess a remarkable regenerative potential that has long been investigated to resolve these issues. Some breakthroughs have been achieved thus far to replace the damaged brain cells; however, the procedure's invasiveness has prompted scientists to investigate using stem-cell small extracellular vesicles (sEVs) as a non-invasive cell-free therapy to address the limitations of cell therapy. With the advancement of technology to understand the molecular changes of neurodegenerative diseases, efforts have been made to enrich stem cells' sEVs with miRNAs to increase the therapeutic efficacy of the sEVs. In this article, the pathophysiology of various neurodegenerative diseases is highlighted. The role of miRNAs from sEVs as biomarkers and treatments is also discussed. Lastly, the applications and delivery of stem cells and their miRNA-enriched sEVs for treating neurodegenerative diseases are emphasised and reviewed. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
21. Homing of placenta-derived mesenchymal stem cells after perinatal intracerebral transplantation in a rat model.
- Author
-
Schoeberlein A, Mueller M, Reinhart U, Sager R, Messerli M, and Surbek DV
- Subjects
ANIMAL experimentation ,CEREBRAL ventricles ,CELL physiology ,CELL motility ,NEURONS ,RATS ,STEM cells - Abstract
OBJECTIVE: The aim of this study is to assess early homing of placenta-derived stem cells after perinatal intracerebral transplantation in rats. STUDY DESIGN: Neonatal Wistar rats (2-4 days old) were anesthetized, and 250,000 human placenta-derived mesenchymal stem cells (MSC) injected into the lateral ventricle or the paraventricular white matter using a stereotactic frame. Donor MSC were detected by immunohistochemistry using an antihuman HLA-ABC antibody. RESULTS: In all, 84% of the animals survived the transplantation. Donor cells were detected in the brain ventricle 1-2 hours posttransplantation. After 4 hours, donor cells migrated throughout the ventricular system. At 1-4 weeks after transplantation, some cells had migrated into the periventricular white matter. CONCLUSION: Human placenta-derived MSC were successfully transplanted into the lateral ventricles of neonatal rats. Donor cells survived, homed, and migrated in the recipient brains. Proliferation and differentiation analysis and functional tests will assess the therapeutic effects of stem cell transplantation. [ABSTRACT FROM AUTHOR]
- Published
- 2011
22. Effect of 3D Spheroid Culturing on NF-κB Signaling Pathway and Neurogenic Potential in Human Amniotic Fluid Stem Cells.
- Author
-
Valiulienė, Giedrė, Zentelytė, Aistė, Beržanskytė, Elizabet, and Navakauskienė, Rūta
- Subjects
AMNIOTIC liquid ,STEM cells ,EXTRACELLULAR matrix proteins ,CELLULAR signal transduction ,GENE expression ,SOMATIC cells - Abstract
Human amniotic fluid stem cells (hAFSCs) are known for their advantageous properties when compared to somatic stem cells from other sources. Recently hAFSCs have gained attention for their neurogenic potential and secretory profile. However, hAFSCs in three-dimensional (3D) cultures remain poorly investigated. Therefore, we aimed to evaluate cellular properties, neural differentiation, and gene and protein expression in 3D spheroid cultures of hAFSCs in comparison to traditional two-dimensional (2D) monolayer cultures. For this purpose, hAFSCs were obtained from amniotic fluid of healthy pregnancies and cultivated in vitro, either in 2D, or 3D under untreated or neuro-differentiated conditions. We observed upregulated expression of pluripotency genes OCT4, NANOG, and MSI1 as well as augmentation in gene expression of NF-κB−TNFα pathway genes (NFKB2, RELA and TNFR2), associated miRNAs (miR103a-5p, miR199a-3p and miR223-3p), and NF-κB p65 protein levels in untreated hAFSC 3D cultures. Additionally, MS analysis of the 3D hAFSCs secretome revealed protein upregulation of IGFs signaling the cascade and downregulation of extracellular matrix proteins, whereas neural differentiation of hAFSC spheroids increased the expression of SOX2, miR223-3p, and MSI1. Summarizing, our study provides novel insights into how 3D culture affects neurogenic potential and signaling pathways of hAFSCs, especially NF-κB, although further studies are needed to elucidate the benefits of 3D cultures more thoroughly. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
23. Cytochalasin B Influences Cytoskeletal Organization and Osteogenic Potential of Human Wharton's Jelly Mesenchymal Stem Cells.
- Author
-
Pampanella, Luca, Abruzzo, Provvidenza Maria, Tassinari, Riccardo, Alessandrini, Andrea, Petrocelli, Giovannamaria, Ragazzini, Gregorio, Cavallini, Claudia, Pizzuti, Valeria, Collura, Nicoletta, Canaider, Silvia, Facchin, Federica, and Ventura, Carlo
- Subjects
MESENCHYMAL stem cells ,CYTOLOGY ,JELLY ,STEM cells ,UMBILICAL cord ,REGENERATIVE medicine - Abstract
Among perinatal stem cells of the umbilical cord, human Wharton's jelly mesenchymal stem cells (hWJ-MSCs) are of great interest for cell-based therapy approaches in regenerative medicine, showing some advantages over other MSCs. In fact, hWJ-MSCs, placed between embryonic and adult MSCs, are not tumorigenic and are harvested with few ethical concerns. Furthermore, these cells can be easily cultured in vitro, maintaining both stem properties and a high proliferative rate for several passages, as well as trilineage capacity of differentiation. Recently, it has been demonstrated that cytoskeletal organization influences stem cell biology. Among molecules able to modulate its dynamics, Cytochalasin B (CB), a cyto-permeable mycotoxin, influences actin microfilament polymerization, thus affecting several cell properties, such as the ability of MSCs to differentiate towards a specific commitment. Here, we investigated for the first time the effects of a 24 h-treatment with CB at different concentrations (0.1–3 μM) on hWJ-MSCs. CB influenced the cytoskeletal organization in a dose-dependent manner, inducing changes in cell number, proliferation, shape, and nanomechanical properties, thus promoting the osteogenic commitment of hWJ-MSCs, as confirmed by the expression analysis of osteogenic/autophagy markers. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
24. Are We Getting It Right? A Scoping Review of Outcomes Reported in Cell Therapy Clinical Studies for Cerebral Palsy.
- Author
-
Finch-Edmondson, Megan, Paton, Madison C. B., Honan, Ingrid, Karlsson, Petra, Stephenson, Candice, Chiu, Darryl, Reedman, Sarah, Griffin, Alexandra R., Morgan, Catherine, and Novak, Iona
- Subjects
CELLULAR therapy ,CEREBRAL palsy ,FAMILY values ,COMMUNITIES ,QUALITY of life - Abstract
Cell therapies are an emergent treatment for cerebral palsy (CP) with promising evidence demonstrating efficacy for improving gross motor function. However, families value improvements in a range of domains following intervention and the non-motor symptoms, comorbidities and complications of CP can potentially be targeted by cell therapies. We conducted a scoping review to describe all outcomes that have been reported in cell therapy studies for CP to date, and to examine what instruments were used to capture these. Through a systematic search we identified 54 studies comprising 2066 participants that were treated with a range of cell therapy interventions. We categorized the reported 53 unique outcome instruments and additional descriptive measures into 10 categories and 12 sub-categories. Movement and Posture was the most frequently reported outcome category, followed by Safety, however Quality of Life, and various prevalent comorbidities and complications of CP were infrequently reported. Notably, many outcome instruments used do not have evaluative properties and thus are not suitable for measuring change following intervention. We provide a number of recommendations to ensure that future trials generate high-quality outcome data that is aligned with the priorities of the CP community. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
25. The potential of cell therapies for cerebral palsy: where are we today?
- Author
-
Novak, Iona, Paton, Madison CB, Griffin, Alexandra R, Jackman, Michelle, Blatch-Williams, Remy K, and Finch-Edmondson, Megan
- Published
- 2023
- Full Text
- View/download PDF
26. Next generation of neurological therapeutics: Native and bioengineered extracellular vesicles derived from stem cells.
- Author
-
Shilin Jin, Zhongyue Lv, Lin Kang, Jiayi Wang, Chengcheng Tan, Liming Shen, Liang Wang, and Jing Liu
- Subjects
EXTRACELLULAR vesicles ,STEM cells ,BIOENGINEERING ,STEM cell treatment ,CENTRAL nervous system - Abstract
Extracellular vesicles (EVs)-based cell-free therapy, particularly stem cell-derived extracellular vesicles (SC-EVs), offers new insights into treating a series of neurological disorders and becomes a promising candidate for alternative stem cell regenerative therapy. Currently, SC-EVs are considered direct therapeutic agents by themselves and/or dynamic delivery systems as they have a similar regenerative capacity of stem cells to promote neurogenesis and can easily load many functional small molecules to recipient cells in the central nervous system. Meanwhile, as non-living entities, SC-EVs avoid the uncontrollability and manufacturability limitations of live stem cell products in vivo (e.g., low survival rate, immune response, and tumorigenicity) and in vitro (e.g., restricted sources, complex preparation processes, poor quality control, low storage, shipping instability, and ethical controversy) by strict quality control system. Moreover, SC-EVs can be engineered or designed to enhance further overall yield, increase bioactivity, improve targeting, and extend their half-life. Here, this review provides an overview on the biological properties of SC-EVs, and the current progress in the strategies of native or bioengineered SC-EVs for nerve injury repairing is presented. Then we further summarize the challenges of recent research and perspectives for successful clinical application to advance SC-EVs from bench to bedside in neurological diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
27. Different Sourced Extracellular Vesicles and Their Potential Applications in Clinical Treatments.
- Author
-
Bahmani, Leila and Ullah, Mujib
- Subjects
EXTRACELLULAR vesicles ,DRUG delivery systems ,CLINICAL medicine ,ARTIFICIAL cells ,CELL populations - Abstract
Extracellular vesicles (EVs) include a heterogeneous group of natural cell-derived nanostructures that are increasingly regarded as promising biotherapeutic agents and drug delivery vehicles in human medicine. Desirable intrinsic properties of EVs including the ability to bypass natural membranous barriers and to deliver their unique biomolecular cargo to specific cell populations position them as fiercely competitive alternatives for currently available cell therapies and artificial drug delivery platforms. EVs with distinct characteristics can be released from various cell types into the extracellular environment as a means of transmitting bioactive components and altering the status of the target cell. Despite the existence of a large number of preclinical studies confirming the therapeutic efficacy of different originated EVs for treating several pathological conditions, in this review, we first provide a brief overview of EV biophysical properties with an emphasis on their intrinsic therapeutic benefits over cell-based therapies and synthetic delivery systems. Next, we describe in detail different EVs derived from distinct cell sources, compare their advantages and disadvantages, and recapitulate their therapeutic effects on various human disorders to highlight the progress made in harnessing EVs for clinical applications. Finally, knowledge gaps and concrete hurdles that currently hinder the clinical translation of EV therapies are debated with a futuristic perspective. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
28. Exosomes Derived from Human Amniotic Fluid Mesenchymal Stem Cells Preserve Microglia and Neuron Cells from Aβ.
- Author
-
Zavatti, Manuela, Gatti, Martina, Beretti, Francesca, Palumbo, Carla, and Maraldi, Tullia
- Subjects
MICROGLIA ,MESENCHYMAL stem cells ,AMNIOTIC liquid ,EXOSOMES ,STEM cells ,NEURONS - Abstract
Background: Neuroinflammation is involved in neuronal cell death that occurs in neurodegenerative diseases such as Alzheimer's disease (AD). Microglia play important roles in regulating the brain amyloid beta (Aβ) levels, so immunomodulatory properties exerted by mesenchymal stem cells may be exploited to treat this pathology. The evidence suggests that the mechanism of action of human amniotic fluid stem cells (hAFSCs) is through their secretome, which includes exosomes (exo). Methods: We examined the effect of exosomes derived from human amniotic fluid stem cells (hAFSCs-exo) on activated BV-2 microglia cells by lipopolysaccharide (LPS) as a neuroinflammation model. To investigate the exo effect on the interplay between AD neurons and microglia, SH-SY5Y neuroblastoma cells treated with Aβ were exposed to a conditioned medium (CM) obtained from activated BV-2 or co-culture systems. Results: We found that the upregulation of the markers of pro-inflammatory microglia was prevented when exposed to hAFSC-exo whereas the markers of the anti-inflammatory macrophage phenotype were not affected. Interestingly, the hAFSC-exo pretreatment significantly inhibited the oxidative stress rise and apoptosis occurring in the neurons in presence of both microglia and Aβ. Conclusion: We demonstrated that hAFSC-exo mitigated an inflammatory injury caused by microglia and significantly recovered the neurotoxicity, suggesting that hAFSC-exo may be a potential therapeutic agent for inflammation-related neurological conditions, including AD. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
29. Investigation of the Reparative and Regenerative Effects of Human Adipose Tissue Mesenchymal Stem Cells on Epidermal Cells Exposed to UVB Ray.
- Author
-
Arslan, Yasemin Tin and Yenisey, Çiğdem
- Subjects
STEM cells ,ADIPOSE tissues ,KERATINOCYTES ,ULTRAVIOLET radiation ,HUMAN body - Abstract
Copyright of Meandros Medical & Dental Journal is the property of Galenos Yayinevi Tic. LTD. STI and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2022
- Full Text
- View/download PDF
30. The safety and efficacy of umbilical cord blood mononuclear cells in individuals with spastic cerebral palsy: a randomized double-blind sham-controlled clinical trial.
- Author
-
Zarrabi, Morteza, Akbari, Masood Ghahvechi, Amanat, Man, Majmaa, Anahita, Moaiedi, Ali Reza, Montazerlotfelahi, Hadi, Nouri, Masoumeh, Hamidieh, Amir Ali, Badv, Reza Shervin, Karimi, Hossein, Rabbani, Ali, Mohebbi, Ali, Rahimi-Dehgolan, Shahram, Rahimi, Rosa, Dehghan, Ensieh, Vosough, Massoud, Abroun, Saeed, Shamsabadi, Farhad Mahvelati, Tavasoli, Ali Reza, and Alizadeh, Houman
- Abstract
Introduction: The current multi-center, randomized, double-blind study was conducted among children with cerebral palsy (CP) to assess the safety and efficacy of umbilical cord blood mononuclear cell (UCB-MNC). We performed the diffusion tensor imaging to assess the changes in the white matter structure.Methods: Males and females aged 4 to 14 years old with spastic CP were included. Eligible participants were allocated in 4:1 ratio to be in the experimental or control groups; respectively. Individuals who were assigned in UCB-MNC group were tested for human leukocyte antigen (HLA) and fully-matched individuals were treated with UCB-MNCs. A single dose (5 × 106 /kg) UCB-MNCs were administered via intrathecal route in experimental group. The changes in gross motor function measure (GMFM)-66 from baseline to one year after treatment were the primary endpoints. The mean changes in modified Ashworth scale (MAS), pediatric evaluation of disability inventory (PEDI), and CP quality of life (CP-QoL) were also evaluated and compared between groups. The mean changes in fractional anisotropy (FA) and mean diffusivity (MD) of corticospinal tract (CST) and posterior thalamic radiation (PTR) were the secondary endpoints. Adverse events were safety endpoint.Results: There were 72 included individuals (36 cases in each group). The mean GMFM-66 scores increased in experimental group; compared to baseline (+ 9.62; 95%CI: 6.75, 12.49) and control arm (β: 7.10; 95%CI: 2.08, 12.76; Cohen's d: 0.62) and mean MAS reduced in individuals treated with UCB-MNCs compared to the baseline (-0.87; 95%CI: -1.2, -0.54) and control group (β: -0.58; 95%CI: -1.18, -0.11; Cohen's d: 0.36). The mean PEDI scores and mean CP-QoL scores in two domains were higher in the experimental group compared to the control. The imaging data indicated that mean FA increased and MD decreased in participants of UCB-MNC group indicating improvements in white matter structure. Lower back pain, headaches, and irritability were the most common adverse events within 24 h of treatment that were related to lumbar puncture. No side effects were observed during follow-up.Conclusions: This trial showed that intrathecal injection of UCB-MNCs were safe and effective in children with CP.Trial Registration: The study was registered with ClinicalTrials.gov ( NCT03795974 ). [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
31. Retroviral infection of human neurospheres and use of stem Cell EVs to repair cellular damage.
- Author
-
Branscome, Heather, Khatkar, Pooja, Al Sharif, Sarah, Yin, Dezhong, Jacob, Sheela, Cowen, Maria, Kim, Yuriy, Erickson, James, Brantner, Christine A., El-Hage, Nazira, Liotta, Lance A., and Kashanchi, Fatah
- Subjects
RETROVIRUS diseases ,STEM cells ,INDUCED pluripotent stem cells ,EXTRACELLULAR vesicles ,PROGENITOR cells - Abstract
HIV-1 remains an incurable infection that is associated with substantial economic and epidemiologic impacts. HIV-associated neurocognitive disorders (HAND) are commonly linked with HIV-1 infection; despite the development of combination antiretroviral therapy (cART), HAND is still reported to affect at least 50% of HIV-1 infected individuals. It is believed that the over-amplification of inflammatory pathways, along with release of toxic viral proteins from infected cells, are primarily responsible for the neurological damage that is observed in HAND; however, the underlying mechanisms are not well-defined. Therefore, there is an unmet need to develop more physiologically relevant and reliable platforms for studying these pathologies. In recent years, neurospheres derived from induced pluripotent stem cells (iPSCs) have been utilized to model the effects of different neurotropic viruses. Here, we report the generation of neurospheres from iPSC-derived neural progenitor cells (NPCs) and we show that these cultures are permissive to retroviral (e.g. HIV-1, HTLV-1) replication. In addition, we also examine the potential effects of stem cell derived extracellular vesicles (EVs) on HIV-1 damaged cells as there is abundant literature supporting the reparative and regenerative properties of stem cell EVs in the context of various CNS pathologies. Consistent with the literature, our data suggests that stem cell EVs may modulate neuroprotective and anti-inflammatory properties in damaged cells. Collectively, this study demonstrates the feasibility of NPC-derived neurospheres for modeling HIV-1 infection and, subsequently, highlights the potential of stem cell EVs for rescuing cellular damage induced by HIV-1 infection. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
32. Emerging concepts in the treatment of optic neuritis: mesenchymal stem cell-derived extracellular vesicles.
- Author
-
Aneesh, Anagha, Liu, Alice, Moss, Heather E., Feinstein, Douglas, Ravindran, Sriram, Mathew, Biji, and Roth, Steven
- Subjects
NEUROMYELITIS optica ,OPTIC neuritis ,EXTRACELLULAR vesicles ,AUTOIMMUNE diseases ,MESENCHYMAL stem cells ,MULTIPLE sclerosis - Abstract
Background: Optic neuritis (ON) is frequently encountered in multiple sclerosis, neuromyelitis optica spectrum disorder, anti-myelin oligodendrocyte glycoprotein associated disease, and other systemic autoimmune disorders. The hallmarks are an abnormal optic nerve and inflammatory demyelination; episodes of optic neuritis tend to be recurrent, and particularly for neuromyelitis optica spectrum disorder, may result in permanent vision loss. Main Body: Mesenchymal stem cell (MSC) therapy is a promising approach that results in remyelination, neuroprotection of axons, and has demonstrated success in clinical studies in other neuro-degenerative diseases and in animal models of ON. However, cell transplantation has significant disadvantages and complications. Cell-free approaches utilizing extracellular vesicles (EVs) produced by MSCs exhibit anti-inflammatory and neuroprotective effects in multiple animal models of neuro-degenerative diseases and in rodent models of multiple sclerosis (MS). EVs have potential to be an effective cell-free therapy in optic neuritis because of their anti-inflammatory and remyelination stimulating properties, ability to cross the blood brain barrier, and ability to be safely administered without immunosuppression. Conclusion: We review the potential application of MSC EVs as an emerging treatment strategy for optic neuritis by reviewing studies in multiple sclerosis and related disorders, and in neurodegeneration, and discuss the challenges and potential rewards of clinical translation of EVs including cell targeting, carrying of therapeutic microRNAs, and prolonging delivery for treatment of optic neuritis. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
33. Current Status and Challenges of Stem Cell Treatment for Alzheimer's Disease.
- Author
-
Pacheco-Herrero, Mar, Soto-Rojas, Luis O., Reyes-Sabater, Heidy, Garcés-Ramirez, Linda, de la Cruz López, Fidel, Villanueva-Fierro, Ignacio, and Luna-Muñoz, José
- Subjects
ALZHEIMER'S disease ,STEM cell treatment ,PROGRESSIVE supranuclear palsy ,TAU proteins ,AMYLOID plaque - Abstract
Neurodegenerative diseases called tauopathies, such as Alzheimer's disease (AD), frontotemporal dementia, progressive supranuclear palsy, and Parkinson's disease, among others, are characterized by the pathological processing and accumulation of tau protein. AD is the most prevalent neurodegenerative disease and is characterized by two lesions: neurofibrillary tangles (NFTs) and neuritic plaques. The presence of NFTs in the hippocampus and neocortex in early and advanced stages, respectively, correlates with the patient's cognitive deterioration. So far, no drugs can prevent, decrease, or limit neuronal death due to abnormal pathological tau accumulation. Among potential non-pharmacological treatments, physical exercise has been shown to stimulate the development of stem cells (SCs) and may be useful in early stages. However, this does not prevent neuronal death from the massive accumulation of NFTs. In recent years, SCs therapies have emerged as a promising tool to repopulate areas involved in cognition in neurodegenerative diseases. Unfortunately, protocols for SCs therapy are still being developed and the mechanism of action of such therapy remains unclear. In this review, we show the advances and limitations of SCs therapy. Finally, we provide a critical analysis of its clinical use for AD. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
34. 系统评价脐带来源细胞移植治疗脑性瘫痪患者的疗效与安全性.
- Author
-
钟李青, 钟珊珊, 韩伟超, 胡润凯, 何淑芬, and 丁少波
- Subjects
PEOPLE with cerebral palsy ,CLINICAL trial registries ,CELL transplantation ,STEM cell treatment ,CEREBRAL palsy - Abstract
Copyright of Chinese Journal of Tissue Engineering Research / Zhongguo Zuzhi Gongcheng Yanjiu is the property of Chinese Journal of Tissue Engineering Research and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2021
- Full Text
- View/download PDF
35. Stem cell-based therapy as a promising approach in Alzheimer's disease: current perspectives on novel treatment.
- Author
-
Bagheri-Mohammadi, Saeid
- Abstract
Alzheimer's disease (AD) is a neuronal disorder with insidious onset and slow progression, leading to growing global concern with huge implications for individuals and society. The occurrence of AD has been increased and has become an important health issue throughout the world. In recent years, the care of more than 35 million patients with AD costs over $ 600 billion per year, it is approximately 1 percent of the global Gross Domestic Product. Currently, the therapeutic approach is not effective for neurological deficits especially after the development of these major neurological disorders. The discovery of the technique called cell-based therapy has shown promising results and made important conclusions beyond AD using the stem cells approach. Here we review recent progress on stem cell-based therapy in the context of AD. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
36. Stem cells in the treatment of renal fibrosis: a review of preclinical and clinical studies of renal fibrosis pathogenesis.
- Author
-
Liu, Yiping, Su, Yan-Yan, Yang, Qian, and Zhou, Tianbiao
- Subjects
RENAL fibrosis ,STEM cell treatment ,CHRONIC kidney failure ,STEM cells ,EXTRACELLULAR matrix - Abstract
Renal fibrosis commonly leads to glomerulosclerosis and renal interstitial fibrosis and the main pathological basis involves tubular atrophy and the abnormal increase and excessive deposition of extracellular matrix (ECM). Renal fibrosis can progress to chronic kidney disease. Stem cells have multilineage differentiation potential under appropriate conditions and are easy to obtain. At present, there have been some studies showing that stem cells can alleviate the accumulation of ECM and renal fibrosis. However, the sources of stem cells and the types of renal fibrosis or renal fibrosis models used in these studies have differed. In this review, we summarize the pathogenesis (including signaling pathways) of renal fibrosis, and the effect of stem cell therapy on renal fibrosis as described in preclinical and clinical studies. We found that stem cells from various sources have certain effects on improving renal function and alleviating renal fibrosis. However, additional clinical studies should be conducted to confirm this conclusion in the future. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
37. Prenatal transplantation of human amniotic fluid stem cell could improve clinical outcome of type III spinal muscular atrophy in mice.
- Author
-
Shaw, Steven W., Peng, Shao-Yu, Liang, Ching-Chung, Lin, Tzu-Yi, Cheng, Po-Jen, Hsieh, T'sang-T'ang, Chuang, Hao-Yu, De Coppi, Paolo, and David, Anna L.
- Subjects
AMNIOTIC liquid ,STEM cells ,HEALTH outcome assessment ,UTERUS ,FLOW cytometry - Abstract
Spinal muscular atrophy (SMA) is a single gene disorder affecting motor function in uterus. Amniotic fluid is an alternative source of stem cell to ameliorate SMA. Therefore, this study aims to examine the therapeutic potential of Human amniotic fluid stem cell (hAFSC) for SMA. Our SMA model mice were generated by deletion of exon 7 of Smn gene and knock-in of human SMN2. A total of 16 SMA model mice were injected with 1 × 10
5 hAFSC in uterus, and the other 16 mice served as the negative control. Motor function was analyzed by three behavioral tests. Engraftment of hAFSC in organs were assessed by flow cytometry and RNA scope. Frequency of myocytes, neurons and innervated receptors were estimated by staining. With hAFSC transplantation, 15 fetuses survived (93.75% survival) and showed better performance in all motor function tests. Higher engraftment frequency were observed in muscle and liver. Besides, the muscle with hAFSC transplantation expressed much laminin α and PAX-7. Significantly higher frequency of myocytes, neurons and innervated receptors were observed. In our study, hAFSC engrafted on neuromuscular organs and improved cellular and behavioral outcomes of SMA model mice. This fetal therapy could preserve the time window and treat in the uterus. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
38. Stem cells and COVID-19: are the human amniotic cells a new hope for therapies against the SARS-CoV-2 virus?
- Author
-
Riedel, Rodrigo N., Pérez-Pérez, Antonio, Sánchez-Margalet, Víctor, Varone, Cecilia L., and Maymó, Julieta L.
- Subjects
SARS-CoV-2 ,COVID-19 ,STEM cells ,PANDEMICS ,COVID-19 treatment ,MESENCHYMAL stem cells - Abstract
A new coronavirus respiratory disease (COVID-19) caused by the SARS-CoV-2 virus, surprised the entire world, producing social, economic, and health problems. The COVID-19 triggers a lung infection with a multiple proinflammatory cytokine storm in severe patients. Without effective and safe treatments, COVID-19 has killed thousands of people, becoming a pandemic. Stem cells have been suggested as a therapy for lung-related diseases. In particular, mesenchymal stem cells (MSCs) have been successfully tested in some clinical trials in patients with COVID-19. The encouraging results positioned MSCs as a possible cell therapy for COVID-19. The amniotic membrane from the human placenta at term is a valuable stem cell source, including human amniotic epithelial cells (hAECs) and human mesenchymal stromal cells (hAMSCs). Interestingly, amnion cells have immunoregulatory, regenerative, and anti-inflammatory properties. Moreover, hAECs and hAMSCs have been used both in preclinical studies and in clinical trials against respiratory diseases. They have reduced the inflammatory response and restored the pulmonary tissue architecture in lung injury in vivo models. Here, we review the existing data about the stem cells use for COVID-19 treatment, including the ongoing clinical trials. We also consider the non-cellular therapies that are being applied. Finally, we discuss the human amniotic membrane cells use in patients who suffer from immune/inflammatory lung diseases and hypothesize their possible use as a successful treatment against COVID-19. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
39. Stem Cell Therapy for Pediatric Traumatic Brain Injury.
- Author
-
Lengel, Dana, Sevilla, Cruz, Romm, Zoe L., Huh, Jimmy W., and Raghupathi, Ramesh
- Subjects
BRAIN injuries ,STEM cell treatment ,PEDIATRIC therapy ,STEM cell transplantation ,WHITE matter (Nerve tissue) - Abstract
There has been a growing interest in the potential of stem cell transplantation as therapy for pediatric brain injuries. Studies in pre-clinical models of pediatric brain injury such as Traumatic Brain Injury (TBI) and neonatal hypoxia-ischemia (HI) have contributed to our understanding of the roles of endogenous stem cells in repair processes and functional recovery following brain injury, and the effects of exogenous stem cell transplantation on recovery from brain injury. Although only a handful of studies have evaluated these effects in models of pediatric TBI, many studies have evaluated stem cell transplantation therapy in models of neonatal HI which has a considerable overlap of injury pathology with pediatric TBI. In this review, we have summarized data on the effects of stem cell treatments on histopathological and functional outcomes in models of pediatric brain injury. Importantly, we have outlined evidence supporting the potential for stem cell transplantation to mitigate pathology of pediatric TBI including neuroinflammation and white matter injury, and challenges that will need to be addressed to incorporate these therapies to improve functional outcomes following pediatric TBI. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
40. Establishment of a Novel Fetal Growth Restriction Model and Development of a Stem-Cell Therapy Using Umbilical Cord-Derived Mesenchymal Stromal Cells.
- Author
-
Kitase, Yuma, Sato, Yoshiaki, Arai, Sakiko, Onoda, Atsuto, Ueda, Kazuto, Go, Shoji, Mimatsu, Haruka, Jabary, Mahboba, Suzuki, Toshihiko, Ito, Miharu, Saito, Akiko, Hirakawa, Akihiro, Mukai, Takeo, Nagamura-Inoue, Tokiko, Takahashi, Yoshiyuki, Tsuji, Masahiro, and Hayakawa, Masahiro
- Subjects
STROMAL cells ,FETAL development ,TREATMENT effectiveness ,UTERINE artery ,BLOOD flow - Abstract
Fetal growth restriction (FGR) is a major complication of prenatal ischemic/hypoxic exposure and affects 5%–10% of pregnancies. It causes various disorders, including neurodevelopmental disabilities due to chronic hypoxia, circulatory failure, and malnutrition via the placenta, and there is no established treatment. Therefore, the development of treatments is an urgent task. We aimed to develop a new FGR rat model with a gradual restrictive load of uterus/placental blood flow and to evaluate the treatment effect of the administration of umbilical cord-derived mesenchymal stromal cells (UC-MSCs). To create the FGR rat model, we used ameroid constrictors that had titanium on the outer wall and were composed of C-shaped casein with a notch and center hole inside that gradually narrowed upon absorbing water. The ameroid constrictors were attached to bilateral ovarian/uterine arteries on the 17th day of pregnancy to induce chronic mild ischemia, which led to FGR with over 20% bodyweight reduction. After the intravenous administration of 1 × 10
5 UC-MSCs, we confirmed a significant improvement in the UC-MSC group in a negative geotaxis test at 1 week after birth and a rotarod treadmill test at 5 months old. In the immunobiological evaluation, the total number of neurons counted via the stereological counting method was significantly higher in the UC-MSC group than in the vehicle-treated group. These results indicate that the UC-MSCs exerted a treatment effect for neurological impairment in the FGR rats. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
41. Potential use of stem cells for fertility preservation.
- Author
-
Gauthier‐Fisher, A., Kauffman, A., and Librach, C. L.
- Subjects
STEM cells ,MALE reproductive health ,CELL preservation ,FERTILITY preservation ,SPERMATOGENESIS ,INDUCED pluripotent stem cells ,SOMATIC cells - Abstract
Background: Infertility and gonadal dysfunction can result from gonadotoxic therapies, environmental exposures, aging, or genetic conditions. In men, non‐obstructive azoospermia (NOA) results from defects in the spermatogenic process that can be attributed to spermatogonial stem cells (SSC) or their niche, or both. While assisted reproductive technologies and sperm banking can enable fertility preservation (FP) in men of reproductive age who are at risk for infertility, FP for pre‐pubertal patients remains experimental. Therapeutic options for NOA are limited. The rapid advance of stem cell research and of gene editing technologies could enable new FP options for these patients. Induced pluripotent stem cells (iPSC), SSC, and testicular niche cells, as well as mesenchymal stromal cells (aka medicinal signaling cells, MSCs), have been investigated for their potential use in male FP strategies. Objective: Here, we review the benefits and challenges for three types of stem cell‐based approaches under investigation for male FP, focusing on the role that promising sources of MSC derived from human umbilical cord, specifically human umbilical cord perivascular cells (HUCPVC), could fulfill. These approaches are as follows: 1. isolation and ex vivo expansion of autologous SSC for in vivo transplantation or in vitro spermatogenesis; 2. in vitro differentiation toward germ cell and testicular somatic cell lineages using autologous SSC, or stem cells such iPSC or MSC; and 3. protection or regeneration of the spermatogenic niche after gonadotoxic insults in vivo. Conclusion: Our studies suggest that HUCPVC are promising sources of cells that could be utilized in multiple aspects of male FP strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
42. Commentary – Exosomes: Realization of the great therapeutic potential of stem cells.
- Author
-
Volpe, J.J.
- Subjects
CEREBRAL anoxia-ischemia ,STEM cells ,EXOSOMES ,VESICLES (Cytology) ,NEURAL stem cells ,STEM cell transplantation - Published
- 2020
- Full Text
- View/download PDF
43. vسلول های بنیادی مزانشیمی: میانکنش با سلول های ایمنی و ویژگی های سرکوب تعدیل ایمنی
- Author
-
سید مهدی حسینی, فاطمه منتظری, سید مهدی کلانتر, احمدرضا بهرامی, فاطمه زارعین, and مریم مقدم متین
- Subjects
INFLAMMATION prevention ,ADIPOSE tissues ,BONE marrow ,CELL physiology ,CHEMOKINES ,CYTOKINES ,GROWTH factors ,IMMUNE system ,IMMUNITY ,IMMUNOLOGICAL adjuvants ,IMMUNOSUPPRESSION ,MEDICAL specialties & specialists ,REGENERATION (Biology) ,STEM cells ,EXOSOMES - Abstract
Background and Objectives Recently, mesenchymal stem cells have attracted much attention in regenerative medicine and cell-based therapies. Mesenchymal stem cells are used in regenerative medicine mainly based on their capacity to differentiate into several cell lineages, low immunogenicity, and in particular their anti-inflammatory and immunosuppressive-immunomodulatory properties. Materials and Methods The present manuscript, by reviewing more than 150 recent published articles, introduces the latest information regarding the anti-inflammatory and immunosuppressiveimmunomodulatory properties of the mesenchymal stem cells. Results The fibroblast-like mesenchymal stem cells can be isolated from various sources such as bone marrow, adipose tissue, skeletal muscle, Wharton jelly, umbilical cord, placenta, and amniotic fluid. The immunomodulatory properties of mesenchymal stem cells result from their interactions with innate and adaptive immune systems to inhibit immune cells. Such an inhibitory function is due to the interaction of these cells with immune cells through direct cellcell contact or via paracrine secretory factors. The composition of these secretions or secretom of these cells includes various components such as growth factors, cytokines, chemokines, anti-inflammatory mediators and exosomes. Many in vivo studies in animal models have demonstrated that the ability of mesenchymal stem cells to regenerate and repair tissues is more attributed to their immunosuppressive-immunomodulatory function rather than their proliferative properties. Conclusions Therefore, understanding the mechanisms establishing the interactions of these cells with immune system would be important for their use as a promising therapeutic approach in the future for treatment of immunological diseases as well as in the field of regenerative medicine. [ABSTRACT FROM AUTHOR]
- Published
- 2020
44. Human intracerebroventricular (ICV) injection of autologous, non-engineered, adipose-derived stromal vascular fraction (ADSVF) for neurodegenerative disorders: results of a 3-year phase 1 study of 113 injections in 31 patients.
- Author
-
Duma, Christopher, Kopyov, Oleg, Kopyov, Alex, Berman, Mark, Lander, Elliot, Elam, Michael, Arata, Michael, Weiland, David, Cannell, Ruslana, Caraway, Chad, Berman, Sean, Scord, Kristin, Stemler, Lian, Chung, Karlyssa, Khoudari, Samuel, McRory, Rory, Duma, Chace, Farmer, Sawyer, Bravo, Anthony, and Yassa, Christian
- Abstract
We have chosen to test the safety of human intracerebroventricular (ICV) brain injections of autologous non-genetically-modified adipose-derived stromal vascular fraction (ADSVF). In this IRB-approved trial, 24 patients received ICV ADSVF via an implanted reservoir between 5/22/14 and 5/22/17. Seven others were injected via their ventriculo-peritoneal shunts. Ten patients had Alzheimer's disease (AD), 6 had amyotrophic lateral sclerosis (ALS), 6 had progressive multiple sclerosis (MS-P), 6 had Parkinson's "Plus" (PD+), 1 had spinal cord injury, 1 had traumatic brain injury, and 1 had stroke. Median age was 74 (range 41–83). Injections were planned every 2–3 months. Thirty-one patients had 113 injections. Patients received SVF injection volumes of 3.5–20 cc (median:4 cc) containing 4.05 × 10
5 to 6.2 × 107 cells/cc, which contained an average of 8% hematopoietic and 7.5% adipose stem cells. Follow-up ranged from 0 to 36 months (median: 9.2 months). MRIs post injection(s) were unchanged, except for one AD patient whose hippocampal volume increased from < 5th percentile to 48th percentile (NeuroQuant® volumetric MRI). Of the 10 AD patients, 8 were stable or improved in tests of cognition. Two showed improvement in P-tau and ß-amyloid levels. Of the 6 MS-P patients all are stable or improved. Four of 6 ALS patients died of disease progression. Twelve of 111 injections (11%) led to 1-4 days of transient meningismus, and mild temperature elevation, which resolved with acetaminophen and/or dexamethasone. Two (1.8% of injections) required hospitalization for these symptoms. One patient (0.9% of injections) had his reservoir removed and later replaced for presumed infection. In this Phase 1 safety trial, ADSVF was safely injected into the human brain ventricular system in patients with no other treatment options. Secondary endpoints of clinical improvement or stability were particularly promising in the AD and MS-P groups. These results will be submitted for a Phase 2 FDA-approved trial. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
45. Bone Tissue Engineering Using Human Cells: A Comprehensive Review on Recent Trends, Current Prospects, and Recommendations.
- Author
-
Kargozar, Saeid, Mozafari, Masoud, Hamzehlou, Sepideh, Brouki Milan, Peiman, Kim, Hae-Won, and Baino, Francesco
- Subjects
TISSUE engineering ,SOMATIC cells ,STEM cells - Abstract
The use of proper cells for bone tissue engineering remains a major challenge worldwide. Cells play a pivotal role in the repair and regeneration of the bone tissue in vitro and in vivo. Currently, a large number of differentiated (somatic) and undifferentiated (stem) cells have been used for bone reconstruction alone or in combination with different biomaterials and constructs (e.g., scaffolds). Although the results of the cell transplantation without any supporting or adjuvant material have been very effective with regard to bone healing. Recent advances in bone scaffolding are now becoming new players affecting the osteogenic potential of cells. In the present study, we have critically reviewed all the currently used cell sources for bone reconstruction and discussed the new horizons that are opening up in the context of cell-based bone tissue engineering strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
46. Turning placenta into brain: placental mesenchymal stem cells differentiate into neurons and oligodendrocytes
- Subjects
Stem cells ,Neurons ,Health - Abstract
To link to full-text access for this article, visit this link: http://dx.doi.org/10.1016/j.ajog.2009.10.893 Byline: C. Bettina Portmann-Lanz (a), Andreina Schoeberlein (a), Reto Portmann (b), Stefan Mohr (a), Pierre Rollini (a), Ruth Sager (a), Daniel V. Surbek (a) Keywords: mesenchymal stem cells; neurodifferentiation; placenta Abstract: We aimed to induce neural stem (NSC) and progenitor cells (NPC) from human placental tissues. Author Affiliation: (a) Departments of Obstetrics and Clinical Research, University Women's Hospital, Inselspital, Novartis, Basel, Switzerland (b) Bern University Hospital, University of Bern, Bern, and Friedrich Miescher Institute for Biomedical Research, Novartis, Basel, Switzerland Article History: Received 4 May 2009; Revised 14 August 2009; Accepted 29 October 2009 Article Note: (footnote) Cite this article as: Portmann-Lanz CB, Schoeberlein A, Portmann R, et al. Turning placenta into brain: placental mesenchymal stem cells differentiate into neurons and oligodendrocytes. Am J Obstet Gynecol 2010;202:294.e1-11. , This study was supported in part by a Grant from the Eagle Foundation, Crans-pres-Celigny, Switzerland., The first 2 authors contributed equally to this work.
- Published
- 2010
47. La medicina regenerativa: fundamentos y aplicaciones.
- Author
-
Isaza Médico, Carlos A., Julieta, Henao, and Jainer, Aranzazu
- Published
- 2018
48. Neuroprotective Potential and Paracrine Activity of Stromal Vs. Culture-Expanded hMSC Derived from Wharton Jelly under Co-Cultured with Hippocampal Organotypic Slices.
- Author
-
Dabrowska, Sylwia, Sypecka, Joanna, Jablonska, Anna, Strojek, Lukasz, Wielgos, Miroslaw, Domanska-Janik, Krystyna, and Sarnowska, Anna
- Abstract
Regardless of enormous translational progress in stem cell clinical application, our knowledge about biological determinants of transplantation-related protection is still limited. In addition to adequate selection of the cell source well dedicated to a specific disease and optimal standardization of all other pre-transplant procedures, we have decided to focus more attention to the impact of culture time and environment itself on molecular properties and regenerative capacity of cell cultured in vitro. The aim of this investigation was to determine neuroprotection-linked cell phenotypic and functional changes that could spontaneously take place when freshly isolated Wharton’s jelly mesenchymal stem cell (WJ-MSC) undergo standard selection, growth, and spontaneous differentiation throughout passaging in vitro. For determining their neuroprotective potential, we used experimental model of human WJ-MSC co-culture with intact or oxygen-glucose-deprived (OGD) rat organotypic hippocampal culture (OHC). It has been shown that putative molecular mechanisms mediating regenerative interactions between WJ-MSC and OHC slices relies mainly on mesenchymal cell paracrine activity. Interestingly, it has been also found that the strongest protective effect is exerted by the co-culture with freshly isolated umbilical cord tissue fragments and by the first cohort of human mesenchymal stem cells (hMSCs) migrating out of these fragments (passage 0). Culturing of WJ-derived hMSC in well-controlled standard conditions under air atmosphere up to fourth passage caused unexpected decline of neuroprotective cell effectiveness toward OGD-OHC in the co-culture model. This further correlated with substantial changes in the WJ-MSC phenotype, profile of their paracrine activities as well as with the recipient tissue reaction evaluated by changes in the rat-specific neuroprotection-linked gene expression. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
49. Comparison of the paracrine activity of mesenchymal stem cells derived from human umbilical cord, amniotic membrane and adipose tissue.
- Author
-
Dabrowski, Filip A., Burdzinska, Anna, Kulesza, Agnieszka, Sladowska, Anna, Zolocinska, Aleksandra, Gala, Kamila, Paczek, Leszek, and Wielgos, Miroslaw
- Subjects
ADIPOSE tissues ,AMNION ,CELL differentiation ,COLLAGEN ,CYTOKINES ,EPIDERMAL growth factor ,FLOW cytometry ,INTERLEUKINS ,METALLOPROTEINS ,STAINS & staining (Microscopy) ,STEM cells ,TRANSFORMING growth factors-beta ,TUMOR necrosis factors ,UMBILICAL cord ,PHENOTYPES ,VASCULAR endothelial growth factors ,IN vitro studies - Abstract
Aim The study was conducted to investigate secretory activity and define the paracrine potential of mesenchymal stem cells from human umbilical cord and amniotic membrane (UC-MSCs and AM-MSCs, respectively). Methods UC-MSCs ( n = 6) were obtained from tissue explants using an adherent method after two weeks of incubation. AM-MSCs ( n = 6) were obtained by digestion with tripsin and collagenase. MSC phenotype was confirmed in vitro by performing flow cytometry, differentiation assays and vimentin staining. Supernatants were collected after 48 h culturing in serum-free conditions and the following concentrations were determined: epidermal growth factor (EGF), interleukin (IL)-6, IL-10, tumor necrosis factor-α, transforming growth factor-β (TGF-β), vascular endothelial growth factor-α (VEGF-α) and metalloproteinase (MMP) 1, 8 and 13, using multiplex supernatant cytokine assay. Data were compared with adipose tissue derived MSCs (AD-MSCs, n = 6). Results Both UC-MSC and AM-MSC populations were positively identified as MSCs by flow cytometry and differentiation potential into bone, cartilage and adipose tissue. Using a multiple cytokine detection assay, we proved that both UC-MSCs and AM-MSCs show high secretive capacity. However, the secretion profile differed between cells from various sources. UC-MSCs showed significantly higher production of TGF-β and lower production of VEGF-α, compared to AD-MSCs ( P = 0.004) and AM-MSCs ( P = 0.039) and lower levels of EGF ( P = 0005). AM-MSCs showed significantly lower levels of MMP-8 than UC-MSCs ( P = 0.024); however, there was no difference in levels of released cytokines compared to AD-MSCs. Conclusion AM-MSCs show similar IL production as AD-MSCs, while UC-MSCs have a significantly different profile, which suggests diverse biological potential of both cell types for immunomodulative and regenerative therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
50. Perinatal Brain Injury As a Consequence of Preterm Birth and Intrauterine Inflammation: Designing Targeted Stem Cell Therapies.
- Author
-
Paton, Madison C. B., McDonald, Courtney A., Allison, Beth J., Fahey, Michael C., Jenkin, Graham, and Miller, Suzanne L.
- Subjects
BRAIN injuries ,PREMATURE labor ,STEM cell treatment - Abstract
Chorioamnionitis is a major cause of preterm birth and brain injury. Bacterial invasion of the chorion and amnion, and/or the placenta, can lead to a fetal inflammatory response, which in turn has significant adverse consequences for the developing fetal brain. Accordingly, there is a strong causal link between chorioamnionitis, preterm brain injury and the pathogenesis of severe postnatal neurological deficits and cerebral palsy. Currently there are no treatments to protect or repair against brain injury in preterminfants born after pregnancy compromised by intrauterine infection. This review describes the injurious cascade of events in the pretermbrain in response to a severe fetal inflammatory event.We will highlight specific periods of increased vulnerability, and the potential effects of therapeutic intervention with cell-based therapies. Many clinical trials are underway to investigate the efficacy of stem cells to treat patients with cerebral palsy. Stem cells, obtained from umbilical cord tissue and cord blood, normally discarded after birth, are emerging as a safe and potentially effective therapy. It is not yet known, however, which stemcell type(s) are themost efficacious for administration to preterminfants to treat brain injury-mediated inflammation. Individual stem cell populations found in cord blood and tissue, such as mesenchymal stem cells (MSCs) and endothelial progenitor cells (EPCs), have a number of potential benefits that may specifically target preterm inflammatory- induced brain injury. MSCs have strong immunomodulatory potential, protecting against global and local neuroinflammatory cascades triggered during infection to the fetus. EPCs have angiogenic and vascular reparative qualities that make them ideal for neurovascular repair. A combined therapy using both MSCs and EPCs to target inflammation and promote angiogenesis for re-establishment of vital vessel networks is a treatment concept that warrants further investigation. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.