1
|
Michalak-Micka K, Huerlimann N, Ochsenbein-Kölble N, Ehrbar M, Klar AS, Biedermann T, Moehrlen U. Isolation and characterization of human cKIT positive amniotic fluid stem cells obtained from pregnancies with spina bifida. Sci Rep 2025; 15:20008. [PMID: 40481046 PMCID: PMC12144188 DOI: 10.1038/s41598-025-03518-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 05/20/2025] [Indexed: 06/11/2025] Open
Abstract
Myelomeningocele (MMC) and myeloschisis (MS) are considered as the most severe forms of spina bifida aperta characterized by incomplete closure of the neural tube during the first trimester of pregnancy. Open maternal-fetal surgery has been identified as a promising alternative for the repair of MMC/MS. The main goal of this study was the isolation and characterization of stem cells derived from MMC/MS amniotic fluid samples. Human amniotic fluid samples were obtained from pregnant women who underwent surgery for fetal spina bifida repair. We applied fluorescence activated-cell sorting (FACS) to immunoselect stem cells from heterogeneous populations of amniocytes based on cKIT (CD117) expression. The cKIT+ amniocytes were then contrasted with cKIT- and unselected amniocytes in order to characterize the phenotype of these cells. cKIT-expressing amniocytes exhibited spindle-shape morphology, while amniocytes negative for cKIT were round-shaped. qRT-PCR analysis revealed significant upregulation of mean mRNA levels of KIT and CD90 genes and downregulation of CK8 gene in cKIT-expressing amniocytes, compared to the negative and unsorted cells. The expression of pluripotency antigens was comparable in sorted cKIT+ and cKIT- amniocytes and in unselected cells. Our results are of pivotal importance for the future application of amniotic fluid derived stem cells in spina bidfida repair.
Collapse
Affiliation(s)
- Katarzyna Michalak-Micka
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Steinwiesstrasse 75, Zurich, 8032, Switzerland
- Children's Research Center (CRC), University Children's Hospital Zurich, Zurich, Switzerland
| | - Nadine Huerlimann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Steinwiesstrasse 75, Zurich, 8032, Switzerland
- Children's Research Center (CRC), University Children's Hospital Zurich, Zurich, Switzerland
| | - Nicole Ochsenbein-Kölble
- The Zurich Center for Fetal Diagnosis and Therapy, University of Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
- Department of Obstetrics, University Hospital Zurich, Zurich, Switzerland
| | - Martin Ehrbar
- University of Zurich, Zurich, Switzerland
- Department of Obstetrics, University Hospital Zurich, Zurich, Switzerland
| | - Agnes Silvia Klar
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Steinwiesstrasse 75, Zurich, 8032, Switzerland
- Children's Research Center (CRC), University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Steinwiesstrasse 75, Zurich, 8032, Switzerland
- Children's Research Center (CRC), University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Ueli Moehrlen
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Steinwiesstrasse 75, Zurich, 8032, Switzerland.
- Children's Research Center (CRC), University Children's Hospital Zurich, Zurich, Switzerland.
- The Zurich Center for Fetal Diagnosis and Therapy, University of Zurich, Zurich, Switzerland.
- University of Zurich, Zurich, Switzerland.
- Spina Bifida Center, University Children's Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
2
|
Yue F, Zhao Y, Lv Y, Li S, Wang W, Li Y, Wang S, Wang C. Anti-Tumor Effects of Sheep Umbilical Cord Mesenchymal Stem Cells on Melanoma Cells. Int J Mol Sci 2025; 26:426. [PMID: 39796281 PMCID: PMC11720557 DOI: 10.3390/ijms26010426] [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: 11/23/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/13/2025] Open
Abstract
Melanoma is among the most common malignancies and has recently exhibited increased resistance to treatments, resulting in a more aggressive disease course. Mesenchymal stem cells (MSCs) secrete cytokines both in vivo and in vitro, which regulate tumor cell signaling pathways and the tumor microenvironment, thereby influencing tumor progression. This study investigates the anti-melanogenesis effects of sheep umbilical cord mesenchymal stem cells (SUCMSCs) to assess their potential application in melanoma treatment. Our findings indicate that, in vitro, SUCMSCs reduce melanin content and tyrosinase activity, inhibit melanoma cell viability, proliferation, migration, and invasion, and promote melanoma cell apoptosis. Subsequent in vivo experiments confirmed that SUCMSCs effectively suppress tumor growth, and histological analysis via HE staining revealed notable differences. Additionally, transcriptome sequencing analysis indicated that the anti-tumor effects were primarily mediated through autophagy, apoptosis, and the TGF-β and NF-κB signaling pathways. The RT-qPCR validation results aligned with the transcriptome data. In summary, SUCMSCs exert anti-melanogenesis effects through the interaction of multiple signaling pathways and cytokines, demonstrating significant potential for melanoma treatment.
Collapse
Affiliation(s)
- Fengjiao Yue
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (F.Y.); (Y.Z.); (Y.L.); (S.L.); (W.W.); (Y.L.)
| | - Yuqing Zhao
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (F.Y.); (Y.Z.); (Y.L.); (S.L.); (W.W.); (Y.L.)
| | - Yiting Lv
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (F.Y.); (Y.Z.); (Y.L.); (S.L.); (W.W.); (Y.L.)
| | - Songmei Li
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (F.Y.); (Y.Z.); (Y.L.); (S.L.); (W.W.); (Y.L.)
| | - Weihai Wang
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (F.Y.); (Y.Z.); (Y.L.); (S.L.); (W.W.); (Y.L.)
| | - Yajun Li
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (F.Y.); (Y.Z.); (Y.L.); (S.L.); (W.W.); (Y.L.)
| | - Shujie Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Chunsheng Wang
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (F.Y.); (Y.Z.); (Y.L.); (S.L.); (W.W.); (Y.L.)
| |
Collapse
|
3
|
Athiel Y, Jouannic JM, Lépine M, Maillet C, de Saint Denis T, Larghero J, Guilbaud L. Role of Amniotic Fluid Toxicity in the Pathophysiology of Myelomeningocele: A Narrative Literature Review. Prenat Diagn 2024; 44:1530-1535. [PMID: 39370541 DOI: 10.1002/pd.6681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/06/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
Myelomeningocele is a birth defect whose clinical manifestations are due both to incomplete neural tube closure and the progressive destruction of exposed neuroepithelium during pregnancy. Two hypotheses have been formulated to explain the spinal cord damage in utero: mechanical trauma and chemical factors. The objective of this review was to summarize the current insights about the potential role of amniotic fluid in spinal cord damage in myelomeningocele. Numerous histological and clinical data on animals and humans strongly suggest a progressive degeneration of neural tissue including loss of neural cells, astrogliosis, inflammation, and loss of normal architecture. However, few data have been published about the direct toxicity of amniotic fluid in this neural degeneration, including the potentially toxic effect of meconium. Finally, the chemical and cellular modifications of amniotic fluid composition in myelomeningocele could reflect both the process (toxic effect of meconium) and the consequences of neuroepithelium destruction (release of neural cells). Fetal surgery not only stops the leakage of the cerebrospinal fluid but also reduces the toxic effect of amniotic fluid by restoring the intrauterine environment. Identification of amniotic fluid neurotoxic factors could lead to the development of therapeutic agents designed to protect spinal tissue and improve fetal myelomeningocele outcomes.
Collapse
Affiliation(s)
- Yoann Athiel
- Department of Fetal Medicine, Armand Trousseau Hospital, AP-HP, DMU ORIGYNE, National Reference Center for Rare Disease: Spin@, Sorbonne University, Paris, France
- Stem Cell Biotechnologies Unit, INSERM 976, CIC-BT, AP-HP, Saint-Louis Hospital, Université Paris Cité, Paris, France
| | - Jean-Marie Jouannic
- Department of Fetal Medicine, Armand Trousseau Hospital, AP-HP, DMU ORIGYNE, National Reference Center for Rare Disease: Spin@, Sorbonne University, Paris, France
- Working Group Spina Bifida and Other Dysraphisms, European Reference Network ITHACA, Paris, France
| | - Matthieu Lépine
- Stem Cell Biotechnologies Unit, INSERM 976, CIC-BT, AP-HP, Saint-Louis Hospital, Université Paris Cité, Paris, France
| | - Corentin Maillet
- Department of Fetal Medicine, Armand Trousseau Hospital, AP-HP, DMU ORIGYNE, National Reference Center for Rare Disease: Spin@, Sorbonne University, Paris, France
- Stem Cell Biotechnologies Unit, INSERM 976, CIC-BT, AP-HP, Saint-Louis Hospital, Université Paris Cité, Paris, France
| | - Timothée de Saint Denis
- Department of Pediatric Orthopedic and Reconstructive Surgery, Armand Trousseau Hospital, AP-HP, Sorbonne University, Paris, France
| | - Jérôme Larghero
- Stem Cell Biotechnologies Unit, INSERM 976, CIC-BT, AP-HP, Saint-Louis Hospital, Université Paris Cité, Paris, France
| | - Lucie Guilbaud
- Department of Fetal Medicine, Armand Trousseau Hospital, AP-HP, DMU ORIGYNE, National Reference Center for Rare Disease: Spin@, Sorbonne University, Paris, France
- Stem Cell Biotechnologies Unit, INSERM 976, CIC-BT, AP-HP, Saint-Louis Hospital, Université Paris Cité, Paris, France
- Working Group Spina Bifida and Other Dysraphisms, European Reference Network ITHACA, Paris, France
| |
Collapse
|
4
|
Athiel Y, Jouannic JM, Mauffré V, Dehan C, Adam C, Blot S, Lallemant P, De Saint Denis T, Larghero J, Nasone J, Guilbaud L. Allogenic umbilical cord-derived mesenchymal stromal cells improve motor function in prenatal surgical repair of myelomeningocele: An ovine model study. BJOG 2024; 131:759-767. [PMID: 37492999 DOI: 10.1111/1471-0528.17624] [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: 03/30/2023] [Revised: 07/05/2023] [Accepted: 07/11/2023] [Indexed: 07/27/2023]
Abstract
OBJECTIVE To investigate the effects of an adjuvant allogenic umbilical cord mesenchymal stromal cell (UC-MSC) patch applied during fetal surgery on motor and sphincter function in the ovine MMC model. DESIGN MMC defects were surgically created at 75 days of gestation and repaired 14 days later. POPULATION Ovine MMC model: fetal lambs. METHODS We compared lambs that received a UC-MSC patch with a control group of lambs that received an acellular patch. MAIN OUTCOME MEASURES Clinical neurological assessment was performed at 2 and 24 hours of life and included determination of the Sheep Locomotor Rating scale (SLR), which has been validated in the ovine MMC model. Electrophysical examinations, spine scans and histological analyses were also performed. RESULTS Of the 13 operated lambs, nine were born alive: five had of these had received a UC-MSC patch and four an acellular patch. At 24 hours of life, lambs in the UC-MSC group had a significantly higher score (14 versus 5, P = 0.04). Amyotrophy was significantly more common in the control group (75% versus 0%, P = 0.02). All the lambs in the control group and none of those in the UC-MSC group were incontinent. No significant differences were observed between the UC-MSC and control groups in terms of the presence of spontaneous EMG activity, nerve conduction or spinal evoked potentials. In the microscopic examination, lambs in the UC-MSC group had less fibrosis between the spinal cord and the dermis (mean thickness, 453 versus 3921 μm, P = 0.03) and around the spinal cord (mean thickness, 47 versus 158 μm, P < 0.001). Examination of the spinal cord in the area of the MMC defect showed a higher large neuron density in the UC-MSC group (14.5 versus 5.6 neurons/mm2, P < 0.001). No tumours were observed. CONCLUSIONS Fetal repair of MMC using UC-MSC patches improves motor and sphincter function as well as spinal preservation and reduction of fibrosis.
Collapse
Affiliation(s)
- Yoann Athiel
- Université Paris Cité, INSERM, U976 et Centre d'Investigation Clinique en Biothérapies CIC-BT CBT501, INSERM, Paris, France
- Service de médecine fœtale, APHP, Hôpital Trousseau, DMU ORIGYNE, Sorbonne Université, Paris, France
| | - Jean-Marie Jouannic
- Université Paris Cité, INSERM, U976 et Centre d'Investigation Clinique en Biothérapies CIC-BT CBT501, INSERM, Paris, France
- Service de médecine fœtale, APHP, Hôpital Trousseau, DMU ORIGYNE, Sorbonne Université, Paris, France
- Working Group Spina Bifida and Other Dysraphisms, European Reference Network ITHACA, Paris, France
| | - Vincent Mauffré
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- École Nationale Vétérinaire d'Alfort, BREED, Maison-Alfort, France
| | - Coralie Dehan
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- École Nationale Vétérinaire d'Alfort, BREED, Maison-Alfort, France
| | - Clovis Adam
- Service d'anatomopathologie, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Stéphane Blot
- U955-IMRB, Inserm, École Nationale Vétérinaire d'Alfort, Unité de Neurologie, Maisons-Alfort, France
| | - Pauline Lallemant
- National Reference Center for Rare Disease: Vertebral and Spinal Cord Anomalies (MAVEM Center), AP-HP, Trousseau Hospital, Paris, France
- Sorbonne University, AP-HP, Trousseau Hospital, Paris, France
| | - Timothé De Saint Denis
- Service de Neurochirurgie Pédiatrique, Centre de Référence Chiari, Syringomyélie et Malformations du Rachis et de la Moelle C-MAVEM, et Centre de Référence des Malformations Craniofaciales-CRMR, Paris, France
| | - Jérôme Larghero
- Université Paris Cité, INSERM, U976 et Centre d'Investigation Clinique en Biothérapies CIC-BT CBT501, INSERM, Paris, France
- Unité de Thérapie Cellulaire et Centre MEARY de Thérapie Cellulaire et Génique, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Justine Nasone
- Université Paris Cité, INSERM, U976 et Centre d'Investigation Clinique en Biothérapies CIC-BT CBT501, INSERM, Paris, France
- Unité de Thérapie Cellulaire et Centre MEARY de Thérapie Cellulaire et Génique, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Lucie Guilbaud
- Université Paris Cité, INSERM, U976 et Centre d'Investigation Clinique en Biothérapies CIC-BT CBT501, INSERM, Paris, France
- Service de médecine fœtale, APHP, Hôpital Trousseau, DMU ORIGYNE, Sorbonne Université, Paris, France
| |
Collapse
|
5
|
Lee SY, Du Y, Hassan AES, Brown E, Saadai P, Hirose S, Wang A, Farmer DL. Evolution and Variations of the Ovine Model of Spina Bifida. Fetal Diagn Ther 2023; 50:491-500. [PMID: 37393899 PMCID: PMC10757987 DOI: 10.1159/000531750] [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: 03/04/2023] [Accepted: 06/08/2023] [Indexed: 07/04/2023]
Abstract
Spina bifida is the most common congenital anomaly of the central nervous system and the first non-fatal fetal lesions to be addressed by fetal intervention. While research in spina bifida has been performed in rodent, nonhuman primate, and canine models, sheep have been a model organism for the disease. This review summarizes the history of development of the ovine model of spina bifida, previous applications, and translation into clinical studies. Initially used by Meuli et al. [Nat Med. 1995;1(4):342-7], fetal myelomeningocele defect creation and in utero repair demonstrated motor function preservation. The addition of myelotomy in this model can reproduce hindbrain herniation malformations, which is the leading cause of mortality and morbidity in humans. Since inception, the ovine models have been validated numerous times as the ideal large animal model for fetal repair, with both locomotive scoring and spina bifida defect scoring adding to the rigor of this model. The ovine model has been used to study different methods of myelomeningocele defect repair, the application of various tissue engineering techniques for neuroprotection and bowel and bladder function. The results of these large animal studies have been translated into human clinical trials including Management of Meningocele Study (MOMS) trial that established current standard of care for prenatal repair of spina bifida defects, and the ongoing trials including the Cellular Therapy for In Utero Repair of Myelomeningocele (CuRe) trial using a stem cell patch for repair. The advancement of these life savings and life-altering therapies began in sheep models, and this notable model continues to be used to further the field including current work with stem cell therapy.
Collapse
Affiliation(s)
- Su Yeon Lee
- Division of Pediatric General, Thoracic and Fetal Surgery, University of California Davis Medical Center, Sacramento, California, USA,
- Center for Surgical Bioengineering, University of California Davis, Sacramento, California, USA,
| | - Yimeng Du
- University of California Davis School of Medicine, Sacramento, California, USA
| | - Abd-Elrahman Said Hassan
- Division of Pediatric General, Thoracic and Fetal Surgery, University of California Davis Medical Center, Sacramento, California, USA
- Center for Surgical Bioengineering, University of California Davis, Sacramento, California, USA
| | - Erin Brown
- Division of Pediatric General, Thoracic and Fetal Surgery, University of California Davis Medical Center, Sacramento, California, USA
| | - Payam Saadai
- Division of Pediatric General, Thoracic and Fetal Surgery, University of California Davis Medical Center, Sacramento, California, USA
| | - Shinjiro Hirose
- Division of Pediatric General, Thoracic and Fetal Surgery, University of California Davis Medical Center, Sacramento, California, USA
| | - Aijun Wang
- Center for Surgical Bioengineering, University of California Davis, Sacramento, California, USA
| | - Diana L Farmer
- Division of Pediatric General, Thoracic and Fetal Surgery, University of California Davis Medical Center, Sacramento, California, USA
- Center for Surgical Bioengineering, University of California Davis, Sacramento, California, USA
| |
Collapse
|
6
|
Damianos A, Sammour I. Barriers in translating stem cell therapies for neonatal diseases. Semin Perinatol 2023; 47:151731. [PMID: 36990922 DOI: 10.1016/j.semperi.2023.151731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Over the last 20 years, stem cells of varying origin and their associated secretome have been investigated as a therapeutic option for a myriad of neonatal models of disease, with very promising results. Despite the devastating nature of some of these disorders, translation of the preclinical evidence to the bedside has been slow. In this review, we explore the existing clinical evidence for stem cell therapies in neonates, highlight the barriers faced by researchers and suggest potential solutions to move the field forward.
Collapse
Affiliation(s)
- Andreas Damianos
- Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, Ohio
| | - Ibrahim Sammour
- Riley Hospital for Children, Indiana University, Indianapolis, USA.
| |
Collapse
|
7
|
Kunpalin Y, Vergote S, Joyeux L, Telli O, David AL, Belfort M, De Coppi P, Deprest J. Local host response of commercially available dural patches for fetal repair of spina bifida aperta in rabbit model. Prenat Diagn 2023; 43:370-381. [PMID: 36650109 DOI: 10.1002/pd.6315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/05/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Fetal surgery for spina bifida aperta (SBA) by open hysterotomy typically repairs anatomical native tissue in layers. Increasingly, fetoscopic repair is performed using a dural patch followed by skin closure. We studied the host response to selected commercially available patches currently being used in a fetal rabbit model for spina bifida repair. METHODS SBA was surgically induced at 23-24 days of gestation (term = 31 days). Fetal rabbits were assigned to unrepaired (SBA group), or immediate repair with Duragen™ or Durepair™. Non-operated littermates served as normal controls. At term, spinal cords underwent immunohistochemical staining including Nissl and glial fibrillary acidic protein. We hypothesized that spinal cord coverage with a dural patch and skin closure would preserve motor neuron density within the non-inferiority limit of 201.65 cells/mm2 and reduce inflammation compared to unrepaired SBA fetuses. RESULTS Motor neuron density assessed by Nissl staining was conserved both by Duragen (n = 6, 89.5; 95% CI -158.3 to -20.6) and Durepair (n = 6, 37.0; 95% CI -132.6 to -58.5), whereas density of GFAP-positive cells to quantify inflammation was lower than in unrepaired SBA-fetuses (SBA 2366.0 ± 669.7 cells/mm2 vs. Duragen 1274.0 ± 157.2 cells/mm2 ; p = 0.0002, Durepair 1069.0 ± 270.7 cells/mm2 ; p < 0.0001). CONCLUSIONS Covering the rabbit spinal cord with either Duragen or Durepair followed by skin closure preserves motor neuron density and reduces the inflammatory response.
Collapse
Affiliation(s)
- Yada Kunpalin
- Department of Development and Regeneration, Cluster Woman and Child, Biomedical Sciences, MyFetUZ Fetal Research Center, KU Leuven, Leuven, Belgium.,Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, UK
| | - Simen Vergote
- Department of Development and Regeneration, Cluster Woman and Child, Biomedical Sciences, MyFetUZ Fetal Research Center, KU Leuven, Leuven, Belgium.,Department of Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium
| | - Luc Joyeux
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Onur Telli
- Department of Development and Regeneration, Cluster Woman and Child, Biomedical Sciences, MyFetUZ Fetal Research Center, KU Leuven, Leuven, Belgium
| | - Anna L David
- Department of Development and Regeneration, Cluster Woman and Child, Biomedical Sciences, MyFetUZ Fetal Research Center, KU Leuven, Leuven, Belgium.,Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, UK
| | - Michael Belfort
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Paolo De Coppi
- Department of Development and Regeneration, Cluster Woman and Child, Biomedical Sciences, MyFetUZ Fetal Research Center, KU Leuven, Leuven, Belgium.,Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Jan Deprest
- Department of Development and Regeneration, Cluster Woman and Child, Biomedical Sciences, MyFetUZ Fetal Research Center, KU Leuven, Leuven, Belgium.,Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, UK.,Department of Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium.,Great Ormond Street Institute of Child Health, University College London, London, UK
| |
Collapse
|
8
|
Athiel Y, Jouannic JM, Guilbaud L. [Surgical experimental protocol of fetal myelomeningocele creation and repair in the ovine model (with video)]. GYNECOLOGIE, OBSTETRIQUE, FERTILITE & SENOLOGIE 2022; 50:744-745. [PMID: 35940530 DOI: 10.1016/j.gofs.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/06/2022] [Accepted: 07/27/2022] [Indexed: 06/15/2023]
Affiliation(s)
- Y Athiel
- Service de médecine fœtale, Hôpital Trousseau, Sorbonne Université, APHP, Paris, France; Unité de Thérapie Cellulaire, Biotechnologie des cellules souches, Unité Inserm 976, Paris, France.
| | - J-M Jouannic
- Service de médecine fœtale, Hôpital Trousseau, Sorbonne Université, APHP, Paris, France
| | - L Guilbaud
- Service de médecine fœtale, Hôpital Trousseau, Sorbonne Université, APHP, Paris, France; Unité de Thérapie Cellulaire, Biotechnologie des cellules souches, Unité Inserm 976, Paris, France
| |
Collapse
|
9
|
Athiel Y, Nasone J, Arakelian L, Faivre L, Dugas A, Jouannic JM, Larghero J, Guilbaud L. Biodistribution of allogenic umbilical cord-derived mesenchymal stromal cells after fetal repair of myelomeningocele in an ovine model. Stem Cell Res Ther 2022; 13:300. [PMID: 35841029 PMCID: PMC9284777 DOI: 10.1186/s13287-022-02991-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 06/25/2022] [Indexed: 11/10/2022] Open
Abstract
Background Myelomeningocele (MMC) is a spinal cord congenital defect that leads to paraplegia, sphincter disorders and potential neurocognitive disabilities. Prenatal surgery of MMC provides a significant benefit compared to surgery at birth. Mesenchymal stromal cell (MSC) therapy as an adjuvant treatment for prenatal surgery showed promising results in animal experiments which could be considered for clinical use in human fetuses. Despite numerous reassuring studies on the safety of MSCs administration in humans, no study focused on MSCs biodistribution after a local MSCs graft on the fetal spinal cord. Aim The purpose of our study was to assess the biodistribution of umbilical cord-derived mesenchymal stromal cells (UC-MSCs) at birth in lambs who had a prenatal myelomeningocele repair using a fibrin patch seeded with allogenic UC-MSCs. Methods After isolation, UC-MSCs were tagged using a green fluorescent protein (GFP)-containing lentiviral vector. MMC defects were surgically created at 75 days of gestation and repaired 15 days later using UC-MSCs patch. Lambs were delivered at 142 days and sacrificed. DNA extraction was performed among biopsies of the different organs and q-PCR analysis was used to detect the expression of GFP (GFP DNA coding sequence). Results In our 6 surviving lambs grafted with UC-MSCs, GFP lentivirus genomic DNA was not detected in the organs. Conclusion These reassuring data will support translational application in humans, especially since the first human clinical trial using mesenchymal stromal cells for in-utero treatment of MMC started recently in U.S.A.
Collapse
Affiliation(s)
- Yoann Athiel
- Stem Cell Biotechnologies, U976 et Centre d'Investigation Clinique en Biothérapies CIC-BT CBT501, INSERM, Paris, France.,Department of Fetal Medicine, APHP, Trousseau Hospital, DMU ORIGYNE, Sorbonne University, Paris, France
| | - Justine Nasone
- Stem Cell Biotechnologies, U976 et Centre d'Investigation Clinique en Biothérapies CIC-BT CBT501, INSERM, Paris, France.,Unité de Thérapie Cellulaire et Centre MEARY de Thérapie Cellulaire et Génique, Saint Louis Hospital, Université Paris Cité, Paris, France
| | - Lousineh Arakelian
- Stem Cell Biotechnologies, U976 et Centre d'Investigation Clinique en Biothérapies CIC-BT CBT501, INSERM, Paris, France
| | - Lionel Faivre
- Stem Cell Biotechnologies, U976 et Centre d'Investigation Clinique en Biothérapies CIC-BT CBT501, INSERM, Paris, France.,Unité de Thérapie Cellulaire et Centre MEARY de Thérapie Cellulaire et Génique, Saint Louis Hospital, Université Paris Cité, Paris, France
| | - Anaïs Dugas
- Stem Cell Biotechnologies, U976 et Centre d'Investigation Clinique en Biothérapies CIC-BT CBT501, INSERM, Paris, France.,Department of Fetal Medicine, APHP, Trousseau Hospital, DMU ORIGYNE, Sorbonne University, Paris, France
| | - Jean-Marie Jouannic
- Stem Cell Biotechnologies, U976 et Centre d'Investigation Clinique en Biothérapies CIC-BT CBT501, INSERM, Paris, France.,Department of Fetal Medicine, APHP, Trousseau Hospital, DMU ORIGYNE, Sorbonne University, Paris, France
| | - Jérôme Larghero
- Stem Cell Biotechnologies, U976 et Centre d'Investigation Clinique en Biothérapies CIC-BT CBT501, INSERM, Paris, France.,Unité de Thérapie Cellulaire et Centre MEARY de Thérapie Cellulaire et Génique, Saint Louis Hospital, Université Paris Cité, Paris, France
| | - Lucie Guilbaud
- Stem Cell Biotechnologies, U976 et Centre d'Investigation Clinique en Biothérapies CIC-BT CBT501, INSERM, Paris, France. .,Department of Fetal Medicine, APHP, Trousseau Hospital, DMU ORIGYNE, Sorbonne University, Paris, France.
| |
Collapse
|