1
|
Moghassemi S, Nikanfar S, Dadashzadeh A, Sousa MJ, Wan Y, Sun F, Colson A, De Windt S, Kwaspen L, Kanbar M, Sobhani K, Yang J, Vlieghe H, Li Y, Debiève F, Wyns C, Amorim CA. The revolutionary role of placental derivatives in biomedical research. Bioact Mater 2025; 49:456-485. [PMID: 40177109 PMCID: PMC11964572 DOI: 10.1016/j.bioactmat.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
The human placenta is a transient yet crucial organ that plays a key role in sustaining the relationship between the maternal and fetal organisms. Despite its historical classification as "biowaste," placental tissues have garnered increasing attention since the early 1900s for their significant medical potential, particularly in wound repair and surgical application. As ethical considerations regarding human placental derivatives have largely been assuaged in many countries, they have gained significant attention due to their versatile applications in various biomedical fields, such as biomedical engineering, regenerative medicine, and pharmacology. Moreover, there is a substantial trend toward various animal product substitutions in laboratory research with human placental derivatives, reflecting a broader commitment to advancing ethical and sustainable research methodologies. This review provides a comprehensive examination of the current applications of human placental derivatives, explores the mechanisms behind their therapeutic effects, and outlines the future potential and directions of this rapidly advancing field.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Saba Nikanfar
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Maria João Sousa
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Yuting Wan
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Fengxuan Sun
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Arthur Colson
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Sven De Windt
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Lena Kwaspen
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Marc Kanbar
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Keyvan Sobhani
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jie Yang
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Hanne Vlieghe
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Yongqian Li
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Frédéric Debiève
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christine Wyns
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christiani A. Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
2
|
Vaiasicca S, James DW, Melone G, Saeed O, Francis LW, Corradetti B. Amniotic fluid-derived mesenchymal stem cells as a therapeutic tool against cytokine storm: a comparison with umbilical cord counterparts. Stem Cell Res Ther 2025; 16:151. [PMID: 40156072 PMCID: PMC11951844 DOI: 10.1186/s13287-025-04262-0] [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: 06/06/2024] [Accepted: 03/04/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Several immunosuppressive therapies have been proposed as key treatment options for critically ill patients since the first appearance of severe acute respiratory syndrome coronavirus 2. Mesenchymal stem cells (MSCs) from different sources have been considered for their potential to attenuate the cytokine storm associated to COVID-19 and the consequent multi-organ failure, providing evidence for safe and efficacious treatments. Among them, administration of umbilical cord-derived MSCs (UC-MSCs) has demonstrated a significant increase in survival rates, largely due to their potent immunosuppressive properties. METHODS We applied next-generation sequencing (NGS) analysis to compare the transcriptomic profiles of MSCs isolated from two gestational sources: amniotic fluid (AF) obtained during prenatal diagnosis and their clinically relevant umbilical cord counterparts, for which datasets were publicly available. A full meta-analysis was performed to identify suitable GEO and NGS datasets for comparison between AF- and UC-MSC samples. RESULTS Transcriptome analysis revelaed significant differences between groups, despite both cell lines being strongly involved in the tissue development, crucial to achieve the complex task of wound healing. Significantly enriched hallmark genes suggest AF-MSC superior immunomodulatory features against signaling pathways actively involved in the cytokine storm (i.e., IL-2/STAT, TNF-a/NFkB, IL-2/STAT5, PI3K/AKT/mTOR). CONCLUSIONS The data presented here suggest that AF-MSCs hold significant promise for treating not only COVID-19-associated cytokine storms but also a variety of other inflammatory syndromes (i.e., those induced by bacterial infections, autoimmune disorders, and therapeutic interventions). Realizing the full potential of AF-MSCs as a comprehensive therapeutic approach in inflammatory disease management will require more extensive clinical trials and in-depth mechanistic studies.
Collapse
Affiliation(s)
- Salvatore Vaiasicca
- Advanced Technology Center for Aging Research, IRCCS INRCA, Ancona, Italy
- Department of Life and Environmental Life, Polytechnic University of Marche, Ancona, Italy
| | - David W James
- Centre of NanoHealth, Swansea University Medical School, Swansea, UK
| | - Gianmarco Melone
- Centre of NanoHealth, Swansea University Medical School, Swansea, UK
| | - Omar Saeed
- Centre of NanoHealth, Swansea University Medical School, Swansea, UK
| | - Lewis W Francis
- Centre of NanoHealth, Swansea University Medical School, Swansea, UK
| | - Bruna Corradetti
- Centre of NanoHealth, Swansea University Medical School, Swansea, UK.
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine, Section Oncology/Hematology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
3
|
Chiodelli P, Bonassi Signoroni P, Scalvini E, Farigu S, Giuzzi E, Paini A, Papait A, Stefani FR, Silini AR, Parolini O. Synergistic Effect of Conditioned Medium from Amniotic Membrane Mesenchymal Stromal Cells Combined with Paclitaxel on Ovarian Cancer Cell Viability and Migration in 2D and 3D In Vitro Models. Pharmaceutics 2025; 17:420. [PMID: 40284415 PMCID: PMC12030038 DOI: 10.3390/pharmaceutics17040420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Ovarian cancer accounts for more deaths than any other cancer of the female reproductive system. Despite standard care, recurrence due to tumor spread and chemoresistance is common, highlighting the need for novel therapies. Mesenchymal stromal cells from the human amniotic membrane (hAMSC) and the intact amniotic membrane (hAM) are promising due to their secretion of tumor-modulating bioactive factors, accessibility from biological waste, and ethical favorability. Furthermore, unlike isolated cells, hAM provides an easier, clinically translatable product. We previously demonstrated that hAMSC can inhibit tumor cell proliferation, both in contact and transwell settings, suggesting that hAMSC secrete bioactive factors able to target tumor cells. This study evaluates the anti-tumor effects of bioactive factors from hAMSC and hAM conditioned medium (CM) on ovarian cancer cells in 2D and 3D models, alone or with paclitaxel. Methods: The impact of CM, alone or with paclitaxel, was tested on ovarian cancer cell proliferation, migration, invasion, and on angiogenesis. Results: hAMSC-CM and hAM-CM inhibited the proliferation and migration in 2D cultures and reduced spheroid growth and invasion in 3D models. Combining CM with paclitaxel enhanced anti-tumor effects in both settings. Conclusions: hAMSC-CM and hAM-CM show therapeutic potential against ovarian cancer, with synergistic benefits when combined with paclitaxel.
Collapse
Affiliation(s)
- Paola Chiodelli
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (O.P.)
| | - Patrizia Bonassi Signoroni
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (P.B.S.); (E.S.); (S.F.); (E.G.); (A.P.); (F.R.S.); (A.R.S.)
| | - Elisa Scalvini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (P.B.S.); (E.S.); (S.F.); (E.G.); (A.P.); (F.R.S.); (A.R.S.)
| | - Serafina Farigu
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (P.B.S.); (E.S.); (S.F.); (E.G.); (A.P.); (F.R.S.); (A.R.S.)
| | - Elisabetta Giuzzi
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (P.B.S.); (E.S.); (S.F.); (E.G.); (A.P.); (F.R.S.); (A.R.S.)
| | - Alice Paini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (P.B.S.); (E.S.); (S.F.); (E.G.); (A.P.); (F.R.S.); (A.R.S.)
| | - Andrea Papait
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (O.P.)
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy
| | - Francesca Romana Stefani
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (P.B.S.); (E.S.); (S.F.); (E.G.); (A.P.); (F.R.S.); (A.R.S.)
| | - Antonietta Rosa Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (P.B.S.); (E.S.); (S.F.); (E.G.); (A.P.); (F.R.S.); (A.R.S.)
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (O.P.)
- Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Cappuccini 1, San Giovanni Rotondo, 71013 Foggia, Italy
| |
Collapse
|
4
|
Estrela D, Santos RF, Masserdotti A, Silini A, Parolini O, Pinto IM, Cruz A. Molecular Biomarkers for Timely and Personalized Prediction of Maternal-Fetal Health Risk. Biomolecules 2025; 15:312. [PMID: 40149848 PMCID: PMC11940122 DOI: 10.3390/biom15030312] [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: 11/20/2024] [Revised: 02/14/2025] [Accepted: 02/15/2025] [Indexed: 03/29/2025] Open
Abstract
Molecular biomarker profiling is an emerging field in maternal-fetal health with the potential to transform early detection and prediction of placental dysfunction. By analysing a range of biomarkers in maternal blood, researchers and clinicians can gain crucial insights into placental health, enabling timely interventions to enhance fetal and maternal outcomes. Placental structural function is vital for fetal growth and development, and disruptions can lead to serious pregnancy complications like preeclampsia. While conventional methods such as ultrasound and Doppler velocimetry offer valuable information on fetal growth and blood flow, they have limitations in predicting placental dysfunction before clinical signs manifest. In contrast, molecular biomarker profiling can provide a more comprehensive assessment by measuring proteins, metabolites, and microRNAs (miRNAs) in maternal blood, reflecting the placenta's endocrine and metabolic functions. This approach offers a deeper understanding of placental health and function, aiding in early detection and prediction of complications. Challenges in developing molecular biomarker profiling include pinpointing specific molecular changes in the placenta linked to pathologies, timing predictions of conditions before clinical onset, and understanding how placental dysfunction affects maternal metabolism. Validating specific biomarkers and integrating them effectively into clinical practice requires further research. This review underscores the significance of molecular biomarker profiling as a powerful tool for early detection and prediction of placental dysfunction in maternal-fetal health. Through an exploration of biomarker analysis, we delve into how a deeper understanding of placental health can potentially improve outcomes for both mother and baby. Furthermore, we address the critical need to validate the utility of biomarkers and effectively integrate them into clinical practice.
Collapse
Affiliation(s)
- Daniel Estrela
- International Iberian Nanotechnology Laboratory (INL), 4715-330 Braga, Portugal;
| | - Rita F. Santos
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; (R.F.S.); (I.M.P.)
- Molecular and Analytical Medicine Laboratory, Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Alice Masserdotti
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.M.)
| | - Antonietta Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy;
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.M.)
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00136 Rome, Italy
| | - Inês Mendes Pinto
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; (R.F.S.); (I.M.P.)
- Molecular and Analytical Medicine Laboratory, Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Andrea Cruz
- International Iberian Nanotechnology Laboratory (INL), 4715-330 Braga, Portugal;
| |
Collapse
|
5
|
Galvez P, Ahmed Omar N, Siadous R, Durand M, Comperat L, Lafarge X, Gindraux F, Sentilhes L, Fricain JC, L'Heureux N, Fenelon M. In vitro and in vivo assessment of a new acellular human amnion/chorion membrane device for guided bone regeneration. Sci Rep 2025; 15:5483. [PMID: 39952961 PMCID: PMC11829051 DOI: 10.1038/s41598-025-88814-7] [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: 05/19/2024] [Accepted: 01/31/2025] [Indexed: 02/17/2025] Open
Abstract
Thanks to its unique biological properties, the human amniotic membrane (AM) has shown promising results for guided bone regeneration (GBR), but displays some limitations such as poor space-maintaining ability. This study thus aimed to develop a new amnion/chorion membrane (ACM), with better mechanical properties as well as comparable or improved biological properties for GBR. We first developed a new decellularization method of ACM (DL-ACM) which was validated by DNA staining and quantification, and its cytocompatibility was established in vitro. The thickness of DL-ACM was significantly increased over thirty-fivefold, and its tearing strength and compression strength significantly increased more than tenfold compared to the decellularized AM (DL-AM). In vivo, DL-ACM demonstrated its biocompatibility subcutaneously, and its osteogenic properties were compared to DL-AM and a gold standard membrane in a GBR defect model in rats. Micro-CT and histomorphometric analysis showed that DL-ACM significantly promoted early bone regeneration after 1 week and significantly increased bone regeneration compared to the empty defect and the gold standard membrane over time. In this study, we developed a simple and reproducible method to produce an acellular, non-cytotoxic, and biocompatible DL-ACM. This new membrane is as effective as AM to promote early bone regeneration while demonstrating better biomechanical properties.
Collapse
Affiliation(s)
- Paul Galvez
- Univ. Bordeaux, INSERM, BioTis, U1026, 146 rue Léo Saignat, 33000, Bordeaux, France.
- CHU Bordeaux, Service de Chirurgie Orale, Place Amélie Raba Léon, 33076, Bordeaux, France.
| | - Naïma Ahmed Omar
- Univ. Bordeaux, INSERM, BioTis, U1026, 146 rue Léo Saignat, 33000, Bordeaux, France
| | - Robin Siadous
- Univ. Bordeaux, INSERM, BioTis, U1026, 146 rue Léo Saignat, 33000, Bordeaux, France
| | - Marlène Durand
- Univ. Bordeaux, INSERM, BioTis, U1026, 146 rue Léo Saignat, 33000, Bordeaux, France
- CHU de Bordeaux, CIC 1401, Place Amélie Raba Léon, 33000, Bordeaux, France
- Univ. Bordeaux, INSERM, Institut Bergonié, CIC 1401, 146 rue Léo Saignat, 33000, Bordeaux, France
| | - Léo Comperat
- Univ. Bordeaux, INSERM, BioTis, U1026, 146 rue Léo Saignat, 33000, Bordeaux, France
| | - Xavier Lafarge
- Etablissement Français du Sang Nouvelle-Aquitaine, Laboratoire d'ingénierie Tissulaire et Cellulaire, Place Amélie Raba Léon, 33000, Bordeaux, France
- Univ. Bordeaux, INSERM, U1211, « Maladies Rares : Génétique et Métabolisme », 146 rue Léo Saignat, 33000, Bordeaux, France
| | - Florelle Gindraux
- CHU Besançon, Service de Chirurgie Maxillo-Faciale, Stomatologie et Odontologie Hospitalière, 3 boulevard Alexandre Fleming, 25000, Besançon, France
- Univ. Marie & Louis Pasteur, SINERGIES, 16 route de Gray, 25000, Besançon, France
| | - Loïc Sentilhes
- CHU Bordeaux, Service de Gynécologie-Obstétrique, Place Amélie Raba Léon, 33076, Bordeaux, France
| | - Jean-Christophe Fricain
- Univ. Bordeaux, INSERM, BioTis, U1026, 146 rue Léo Saignat, 33000, Bordeaux, France
- CHU Bordeaux, Service de Chirurgie Orale, Place Amélie Raba Léon, 33076, Bordeaux, France
- CHU Bordeaux, Centre de Compétence des Maladies Rares Orales et Dentaires, O-RARES, Pôle d'odontologie et Santé Buccale, Place Amélie Raba Léon, 33076, Bordeaux, France
| | - Nicolas L'Heureux
- Univ. Bordeaux, INSERM, BioTis, U1026, 146 rue Léo Saignat, 33000, Bordeaux, France
| | - Mathilde Fenelon
- Univ. Bordeaux, INSERM, BioTis, U1026, 146 rue Léo Saignat, 33000, Bordeaux, France
- CHU Bordeaux, Service de Chirurgie Orale, Place Amélie Raba Léon, 33076, Bordeaux, France
- CHU Bordeaux, Centre de Compétence des Maladies Rares Orales et Dentaires, O-RARES, Pôle d'odontologie et Santé Buccale, Place Amélie Raba Léon, 33076, Bordeaux, France
| |
Collapse
|
6
|
Chen Y, Qi W, Wang Z, Niu F. Exosome Source Matters: A Comprehensive Review from the Perspective of Diverse Cellular Origins. Pharmaceutics 2025; 17:147. [PMID: 40006514 PMCID: PMC11858990 DOI: 10.3390/pharmaceutics17020147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/02/2025] [Accepted: 01/14/2025] [Indexed: 02/27/2025] Open
Abstract
Exosomes have emerged as promising therapeutic agents in regenerative medicine. This review introduces a novel cell type-oriented perspective to systematically analyze exosomal properties in regenerative therapies. To our knowledge, this review is the first to comprehensively compare exosomes based on cellular source type, offering unprecedented insights into selecting optimal exosome producers for targeted regenerative applications. Factors beyond cellular origin influencing exosomal therapeutic efficacy, such as donor sites and collection methods, are also explored here. By synthesizing key advances, we propose promising research directions in the end. We aim to accelerate the development of more effective exosome-based regenerative therapies and highlight underexplored directions in this rapidly evolving field.
Collapse
Affiliation(s)
| | | | | | - Feng Niu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 33 Badachu Road, Shijingshan, Beijing 100144, China; (Y.C.)
| |
Collapse
|
7
|
Zhu C, Liu Y, Xu H, Wang S, Zhou H, Cao J, Meng F, Zhang Y. Production of second-generation sheep clones via somatic cell nuclear transfer using amniotic cells as nuclear donors. Theriogenology 2025; 232:79-86. [PMID: 39515062 DOI: 10.1016/j.theriogenology.2024.11.001] [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: 04/17/2024] [Revised: 10/20/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Somatic Cell Nuclear Transfer (SCNT) has transformed animal genetic improvement, gene-editing in model production, xenotransplantation, and conservation efforts for endangered species. However, SCNT-derived embryos occasionally display developmental abnormalities, and following embryo transfer, the miscarriage rate is high. Gene-edited fetuses may experience birth defects, resulting in decreased survival rates. Correct selection of nuclear donor cells is essential for the success of somatic cell cloning. Fibroblasts are the most commonly used cells, but their rapid proliferation increases the risk of genetic mutation, impairing embryo development and production. Conversely, amniotic cells have slower proliferation rates, decreasing the mutation risk during cultivation. Amniotic cells are thus better SCNT candidates than fibroblasts because they offer genomic stability, low tumorigenic and teratogenic risks, reduced immunogenicity, high differentiation potential, ease of accessibility, and fewer ethical concerns. Cells derived from first-generation gene-edited animals exhibit stable genetic structures, reduced susceptibility to genetic alterations and artificial modifications, closely resembling natural cells, and enhanced compatibility with SCNT procedures. Amniotic cells derived from gene-edited sheep fetuses used as nuclear donor cells for SCNT successfully recloned three healthy second-generation gene-edited sheep. Using amniotic cells as nuclear donor cells for SCNT did not significantly alter embryo cleavage rates, blastocyst formation, or fetal birth compared to edited fibroblasts (p > 0.05). However, fetal survival rates were significantly higher than edited fibroblasts (p < 0.05). The results support the potential of amniotic cells as SCNT alternatives, suggesting a promising strategy to improve gene-edited fetus survival rates using first-generation gene-edited sheep-derived amniotic cells.
Collapse
Affiliation(s)
- Chunxiao Zhu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China; Department of Pediatrics, Xinqiao Hospital, Army Medical University, Chongqing, 400073, China; Inner Mongolia Key Laboratory of Biomanufacture, Hohhot, 010018, China
| | - Yiyi Liu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Biomanufacture, Hohhot, 010018, China
| | - Hongyang Xu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Shenyuan Wang
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Biomanufacture, Hohhot, 010018, China
| | - Huanmin Zhou
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Biomanufacture, Hohhot, 010018, China
| | - Junwei Cao
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Biomanufacture, Hohhot, 010018, China.
| | - Fanhuan Meng
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Biomanufacture, Hohhot, 010018, China.
| | - Yanru Zhang
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China; College of Medicine, Hainan Vocational University of Science and Technology, Haikou, 571126, China; Inner Mongolia Key Laboratory of Biomanufacture, Hohhot, 010018, China.
| |
Collapse
|
8
|
Khanabdali R, Shojaee M, Johnson J, Law SQK, Lim MBL, James PF, Tester A, Kalionis B. Profiling the extracellular vesicles of two human placenta-derived mesenchymal stromal cell populations. Exp Cell Res 2025; 444:114387. [PMID: 39706285 DOI: 10.1016/j.yexcr.2024.114387] [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: 08/12/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
Increasing evidence shows extracellular vesicles (EVs) are primarily responsible for the beneficial effects of cell-based therapies. EVs derived from mesenchymal stromal cells (MSCs) show promise as a source of EVs for cell-free therapies. The human placental fetal-maternal interface is a rich and abundant source of MSCs from which EVs can be isolated. This study focusses on chorionic MSCs (CMSC) located on the fetal aspect of the interface and decidual MSCs (DMSC) on the maternal aspect. This study used Ligand-based Exosome Affinity Purification (LEAP) chromatography to isolate EVs from well-characterized placental hTERT-transduced CMSC29 and DMSC23 cell lines, which retain many important stem cell-like properties of primary CMSC and DMSC, respectively. After initial biophysical characterization of the EVs isolated from each cell line, the biological activities and the protein, lipid and small RNA contents of CMSC29-EVs and DMSC23-EVs were compared and assessed. LEAP-purified EVs from both sources were validated at the biophysical level by Spectradyne, Cryo-Transmission Electron Microscopy (Cryo-TEM), and Western blot analysis. EVs from each type were labelled with the live cell stain PKH26 and their in vitro uptake and internalization by human dermal fibroblast cells was assessed, as well as their phosphorylation of the protein kinase B/AKT (AKT) pathway. The protein and lipid contents were analyzed by mass spectrometry and the nucleic acid content by RNA sequencing (RNA-seq). Lastly, the biological activities of the EVs were evaluated in a BioMAP® Diversity PLUS® screen system across a panel of 12 human primary cell-based systems and in vitro cell proliferation. EVs isolated from both DMSC23 and CMSC29 significantly increased proliferation of fibroblasts and showed phosphorylation of the AKT pathway. Protein mass spectrometry analysis identified a large number of proteins including cell surface receptors, cytokines, chemokines, matrix molecules and enzymes in both EV types. Lipidomic analysis identified species including phosphatidylcholine, triacylglycerides and diacylglycerides in both DMSC23 and CMSC29-derived EVs. There were some significant differences in identified microRNAs (miRNAs) between the two EV types. The top differentially expressed miRNAs between the two EV types show pathways association with matrix interaction, transcriptional regulation, proliferation, cellular protein modification processes, and vasculogenesis. Differences were also detected between DMSC23- and CMSC29-EVs in the biological activity they displayed in the BioMAP® Diversity PLUS® screen.
Collapse
Affiliation(s)
- Ramin Khanabdali
- Exopharm Ltd, Level 17, 31 Queen Street, Melbourne, VIC, 3000, Australia
| | - Mozhgan Shojaee
- Exopharm Ltd, Level 17, 31 Queen Street, Melbourne, VIC, 3000, Australia
| | - Jancy Johnson
- Exopharm Ltd, Level 17, 31 Queen Street, Melbourne, VIC, 3000, Australia; University of Melbourne Department of Biochemistry and Pharmacology, Parkville, VIC, 3052, Australia
| | - Sam Q K Law
- Exopharm Ltd, Level 17, 31 Queen Street, Melbourne, VIC, 3000, Australia
| | - Melissa B L Lim
- Exopharm Ltd, Level 17, 31 Queen Street, Melbourne, VIC, 3000, Australia
| | - Patrick F James
- Exopharm Ltd, Level 17, 31 Queen Street, Melbourne, VIC, 3000, Australia
| | - Angus Tester
- Exopharm Ltd, Level 17, 31 Queen Street, Melbourne, VIC, 3000, Australia
| | - Bill Kalionis
- Department of Maternal-Fetal Medicine Pregnancy Research Centre, Royal Women's Hospital, Parkville, VIC, 3052, Australia; University of Melbourne Department of Obstetrics and Gynaecology and Newborn Health, Royal Women's Hospital, Parkville, VIC, 3052, Australia.
| |
Collapse
|
9
|
Fitriani N, Wilar G, Narsa AC, Elamin KM, Wathoni N. Alginate-Based Hydrogels with Amniotic Membrane Stem Cells for Wound Dressing Application. Stem Cells Cloning 2025; 18:1-13. [PMID: 39816853 PMCID: PMC11730520 DOI: 10.2147/sccaa.s493125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 12/07/2024] [Indexed: 01/18/2025] Open
Abstract
Objective Chronic wounds are a common clinical problem that necessitate the exploration of novel regenerative therapies. We report a method to investigate the in vitro wound healing capacity of an innovative biomaterial, which is based on amniotic membrane-derived stem cells (AMSCs) embedded in an alginate hydrogel matrix. The aim of this study was to prepare an sodium alginate-based hydrogel, cross-linked calcium chloride (CaCl2) with the active ingredient AMSC (AMSC/Alg-H) and to evaluate its in vitro effectiveness for wound closure. Methods This hydrogel preparation involved combining sterile solutions of AMSC, sodium alginate, and CaCl2, followed by rinsing with serum-free media. The cells were cultured in different 6-well plates, namely sodium alginate, calcium chloride, AMSC, Alg-H, and AMSC/Alg-H, in complete medium with 10% FBS. The hydrogel was successfully formulated, as confirmed by characterization techniques including Scanning Electron Microscopy (SEM), Fourier Transform Infrared (FTIR) spectroscopy, Differential Scanning Calorimetry (DSC), Cytotoxicity Studies, TGF-β1 Level Measurement by ELISA, and Cell Scratch Wound Assay. Results Cryo-EM characterization of the Alg-H preparation successfully demonstrated the encapsulation of MSCs. FTIR and DSC analyses indicate that crosslinking transpires in Alg-H encapsulating AMSC. The AMSC/Alg-H preparation showed no significant difference in toxicity compared to HaCaT cells (p < 0.05), indicating it was not toxic to HaCaT cells. Furthermore, in the scratch wound assay test at 24 hours, the AMSC/Alg-H preparation achieved 100% wound closure, outperforming both AMSC and Alg-H alone. In vitro assessment revealed that AMSC/Alg-H significantly enhanced key wound healing processes, including cell proliferation and migration, compared to Alg-H. Conclusion Our study demonstrated the promising potential of AMSC/Alg-H as an enhanced regenerative therapy for in vitro wound healing. AMSC/Alg-H was able to maintain the viability of AMSCs and facilitate the formation of tissue-like structures.
Collapse
Affiliation(s)
- Nurul Fitriani
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
- Pharmaceutical Research and Development Laboratory of FARMAKA TROPIS, Faculty of Pharmacy, Universitas Mulawarman, Samarinda, 75119, Indonesia
| | - Gofarana Wilar
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| | - Angga Cipta Narsa
- Pharmaceutical Research and Development Laboratory of FARMAKA TROPIS, Faculty of Pharmacy, Universitas Mulawarman, Samarinda, 75119, Indonesia
| | - Khaled M Elamin
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| |
Collapse
|
10
|
Muntiu A, Papait A, Vincenzoni F, Rossetti DV, Romele P, Cargnoni A, Silini A, Parolini O, Desiderio C. Proteomic analysis of the human amniotic mesenchymal stromal cell secretome by integrated approaches via filter-aided sample preparation. J Proteomics 2025; 310:105339. [PMID: 39448028 DOI: 10.1016/j.jprot.2024.105339] [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: 06/11/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
The immunomodulatory, anti-inflammatory and regenerative properties of the human amniotic mesenchymal stromal cells (hAMSCs) secretome are acknowledged but the understanding of the specific bioactive components remains incomplete. To address these limitations, the present investigation aimed to profile the proteins and peptides content of the hAMSC secretome through sample pretreatment and fractionation on 10 kDa molecular cut-off FASP (Filter Aided Sample Preparation) device and LC-MS analysis. The filter retained protein fraction underwent trypsin digestion, while the unretained was collected unchanged for intact small proteins and peptides analysis. This combined approach (C-FASP) collects in a single step two complementary fractions, advantageously saving sample volume and time of analysis. The bottom-up analysis of the C-FASP proteins fraction >10 kDa confirmed our previous findings, establishing a set of proteins consistently characterizing the hAMSC secretome. The analysis of the fraction <10 kDa, never been investigated to our knowledge, identified peptide fragments of thymosin beta 4 and beta 10, collagen alpha 1 chains I and III, alpha-enolase, and glyceraldehyde-3-phosphate dehydrogenase, involved in wound healing, anti-inflammatory response, tissue repair and regeneration, key biological activities of the secretome. C-FASP provided a comprehensive molecular profile of the hAMSC secretome offering new insights for enhanced therapeutic applications in regenerative medicine. SIGNIFICANCE: In this investigation we originally present the comprehensive proteomic investigation of the human amniotic mesenchymal stromal cell secretome by combining the analysis of the proteome and of the peptidome following sample pretreatment and fractionation by Filter Aided Sample Preparation (FASP) with 10 kDa molecular cut-off in coupling with LC-MS analysis. The proteome fraction retained by FASP filter was analyzed after enzymatic digestion, while the unretained fraction, below 10 kDa molecular mass, was analyzed unchanged in its intact form. This dual approach provides novel insights, previously unexplored, into the molecular components potentially responsible for the immunomodulatory and anti-inflammatory properties of the hAMSC secretome. These findings could significantly enhance the therapeutic potential of hAMSCs in regenerative medicine.
Collapse
Affiliation(s)
- Alexandra Muntiu
- Istituto di Scienze e Tecnologie Chimiche (SCITEC) "Giulio Natta", Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Andrea Papait
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Rome, Italy
| | - Federica Vincenzoni
- Fondazione Policlinico Universitario "Agostino Gemelli" Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Rome, Italy; Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Diana Valeria Rossetti
- Istituto di Scienze e Tecnologie Chimiche (SCITEC) "Giulio Natta", Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Pietro Romele
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Anna Cargnoni
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Antonietta Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Rome, Italy
| | - Claudia Desiderio
- Istituto di Scienze e Tecnologie Chimiche (SCITEC) "Giulio Natta", Consiglio Nazionale delle Ricerche, Rome, Italy.
| |
Collapse
|
11
|
Ge Z, Qiu C, Zhou J, Yang Z, Jiang T, Yuan W, Yu L, Li J. Proteomic analysis of human Wharton's jelly mesenchymal stem/stromal cells and human amniotic epithelial stem cells: a comparison of therapeutic potential. Sci Rep 2024; 14:28061. [PMID: 39543366 PMCID: PMC11564572 DOI: 10.1038/s41598-024-79063-1] [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: 08/15/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
Perinatal stem cells have prominent applications in cell therapy and regenerative medicine. Among them, human Wharton's jelly mesenchymal stem/stromal cells (hWJMSCs) and human amniotic epithelial stem cells (hAESCs) have been widely used. However, the distinction in the therapeutic potential of hWJMSCs and hAESCs is poorly understood. In this study, we reported the phenotypic differences between these two distinct cell types and provided the first systematic comparison of their therapeutic potential in terms of immunomodulation, extracellular matrix (ECM) remodelling, angiogenesis and antioxidative stress using proteomics. The results revealed that the two cell types presented different protein expression profiles and were both promising candidates for cell therapy. Both types of cells demonstrated angiogenic and antifibrotic potential, whereas hAESCs presented superior immunological tolerance and antioxidant properties, which were supported by a series of relevant in vitro assays. Our study provides clues for the selection of appropriate cell types for diverse indications in cell therapy, which contributes to the advancement of their clinical translation and application.
Collapse
Affiliation(s)
- Zhen Ge
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310013, Zhejiang, China
| | - Chen Qiu
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine of Sir Run Run Shaw Hospital, Zhejiang University-Lishui Joint Innovation Center for Life and Health, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, 314400, China
| | - Jiayi Zhou
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine of Sir Run Run Shaw Hospital, Zhejiang University-Lishui Joint Innovation Center for Life and Health, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, 314400, China
| | - Zhuoheng Yang
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine of Sir Run Run Shaw Hospital, Zhejiang University-Lishui Joint Innovation Center for Life and Health, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, 314400, China
| | - Tuoying Jiang
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine of Sir Run Run Shaw Hospital, Zhejiang University-Lishui Joint Innovation Center for Life and Health, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, 314400, China
| | - Weixin Yuan
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, 314400, China
| | - Luyang Yu
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine of Sir Run Run Shaw Hospital, Zhejiang University-Lishui Joint Innovation Center for Life and Health, Zhejiang University, Hangzhou, 310058, China.
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, 314400, China.
| | - Jinying Li
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine of Sir Run Run Shaw Hospital, Zhejiang University-Lishui Joint Innovation Center for Life and Health, Zhejiang University, Hangzhou, 310058, China.
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, 314400, China.
| |
Collapse
|
12
|
Silva-Sousa T, Usuda JN, Al-Arawe N, Frias F, Hinterseher I, Catar R, Luecht C, Riesner K, Hackel A, Schimke LF, Dias HD, Filgueiras IS, Nakaya HI, Camara NOS, Fischer S, Riemekasten G, Ringdén O, Penack O, Winkler T, Duda G, Fonseca DLM, Cabral-Marques O, Moll G. The global evolution and impact of systems biology and artificial intelligence in stem cell research and therapeutics development: a scoping review. Stem Cells 2024; 42:929-944. [PMID: 39230167 DOI: 10.1093/stmcls/sxae054] [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: 06/13/2024] [Accepted: 08/07/2024] [Indexed: 09/05/2024]
Abstract
Advanced bioinformatics analysis, such as systems biology (SysBio) and artificial intelligence (AI) approaches, including machine learning (ML) and deep learning (DL), is increasingly present in stem cell (SC) research. An approximate timeline on these developments and their global impact is still lacking. We conducted a scoping review on the contribution of SysBio and AI analysis to SC research and therapy development based on literature published in PubMed between 2000 and 2024. We identified an 8 to 10-fold increase in research output related to all 3 search terms between 2000 and 2021, with a 10-fold increase in AI-related production since 2010. Use of SysBio and AI still predominates in preclinical basic research with increasing use in clinically oriented translational medicine since 2010. SysBio- and AI-related research was found all over the globe, with SysBio output led by the (US, n = 1487), (UK, n = 1094), Germany (n = 355), The Netherlands (n = 339), Russia (n = 215), and France (n = 149), while for AI-related research the US (n = 853) and UK (n = 258) take a strong lead, followed by Switzerland (n = 69), The Netherlands (n = 37), and Germany (n = 19). The US and UK are most active in SCs publications related to AI/ML and AI/DL. The prominent use of SysBio in ESC research was recently overtaken by prominent use of AI in iPSC and MSC research. This study reveals the global evolution and growing intersection among AI, SysBio, and SC research over the past 2 decades, with substantial growth in all 3 fields and exponential increases in AI-related research in the past decade.
Collapse
Affiliation(s)
- Thayna Silva-Sousa
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, 16816 Neuruppin, Germany
- Fakultät für Gesundheitswissenschaften Brandenburg, Gemeinsame Fakultät der Universität Potsdam, der Medizinischen Hochschule Brandenburg Theodor Fontane, und der Brandenburgischen Technischen Universität Cottbus-Senftenberg, 14476 Potsdam, Germany
| | - Júlia Nakanishi Usuda
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, 16816 Neuruppin, Germany
- Fakultät für Gesundheitswissenschaften Brandenburg, Gemeinsame Fakultät der Universität Potsdam, der Medizinischen Hochschule Brandenburg Theodor Fontane, und der Brandenburgischen Technischen Universität Cottbus-Senftenberg, 14476 Potsdam, Germany
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo (SP), Brazil
| | - Nada Al-Arawe
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, 16816 Neuruppin, Germany
- Fakultät für Gesundheitswissenschaften Brandenburg, Gemeinsame Fakultät der Universität Potsdam, der Medizinischen Hochschule Brandenburg Theodor Fontane, und der Brandenburgischen Technischen Universität Cottbus-Senftenberg, 14476 Potsdam, Germany
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätzsmedizin, 10117 Berlin, Germany
- Department of Hematology, Oncology, and Tumorimmunology, Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Francisca Frias
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, 16816 Neuruppin, Germany
- Fakultät für Gesundheitswissenschaften Brandenburg, Gemeinsame Fakultät der Universität Potsdam, der Medizinischen Hochschule Brandenburg Theodor Fontane, und der Brandenburgischen Technischen Universität Cottbus-Senftenberg, 14476 Potsdam, Germany
| | - Irene Hinterseher
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, 16816 Neuruppin, Germany
- Fakultät für Gesundheitswissenschaften Brandenburg, Gemeinsame Fakultät der Universität Potsdam, der Medizinischen Hochschule Brandenburg Theodor Fontane, und der Brandenburgischen Technischen Universität Cottbus-Senftenberg, 14476 Potsdam, Germany
- Vascular Surgery, Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Rusan Catar
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Christian Luecht
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Katarina Riesner
- Department of Hematology, Oncology, and Tumorimmunology, Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Alexander Hackel
- Clinic for Rheumatology and Clinical Immunology, University Medical Center Schleswig Holstein Campus Lübeck, 23538 Lübeck, Germany
| | - Lena F Schimke
- Department of Immunology, Institute of Biomedical Sciences, USP, SP, Brazil
| | - Haroldo Dutra Dias
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), USP, SP, Brazil
| | | | - Helder I Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo (SP), Brazil
- Department of Medicine, Division of Molecular Medicine, Laboratory of Medical Investigation 29, USP School of Medicine (USPM), São Paulo (SP), Brazil
| | - Niels Olsen Saraiva Camara
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo (SP), Brazil
| | - Stefan Fischer
- Clinic for Rheumatology and Clinical Immunology, University Medical Center Schleswig Holstein Campus Lübeck, 23538 Lübeck, Germany
| | - Gabriela Riemekasten
- Clinic for Rheumatology and Clinical Immunology, University Medical Center Schleswig Holstein Campus Lübeck, 23538 Lübeck, Germany
| | - Olle Ringdén
- Division of Pediatrics, Department of CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Olaf Penack
- Department of Hematology, Oncology, and Tumorimmunology, Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Tobias Winkler
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Georg Duda
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Dennyson Leandro M Fonseca
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), USP, SP, Brazil
| | - Otávio Cabral-Marques
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo (SP), Brazil
- Department of Immunology, Institute of Biomedical Sciences, USP, SP, Brazil
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), USP, SP, Brazil
- Department of Medicine, Division of Molecular Medicine, Laboratory of Medical Investigation 29, USP School of Medicine (USPM), São Paulo (SP), Brazil
- D'OR Institute Research and Education, SP, Brazil
| | - Guido Moll
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätzsmedizin, 10117 Berlin, Germany
| |
Collapse
|
13
|
Heydari P, Mojahedi M, Javaherchi P, Sharifi M, Kharazi AZ. Advances and impact of human amniotic membrane and human amniotic-based materials in wound healing application. Int J Biol Macromol 2024; 281:136596. [PMID: 39419158 DOI: 10.1016/j.ijbiomac.2024.136596] [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: 07/11/2024] [Revised: 10/12/2024] [Accepted: 10/12/2024] [Indexed: 10/19/2024]
Abstract
Wound healing is a complicated process, especially when surgical, traumatic, burn, or pathological injury occurs, which requires different kinds of dressing covers including hydrogels, hydrocolloids, alginates foams and films for treatment. The human amniotic membrane (hAM) is a biodegradable extracellular matrix with unique and tailorable physicochemical and biological properties, generated by the membrane itself or other cells that are located on the membrane surface. It is noted as a promising aid for wound healing and tissue regeneration due to the release of growth factors and cytokines, and its antibacterial and immunosuppressive properties. Moreover, hAM has optimal physical, biological, and mechanical properties, which makes it a much better option as a regenerative skin treatment than existing alternative materials. In addition, this layer has a structure with different layers and cells with different functions, which act as a regenerative geometry and reservoir of bioactive substances and cells for wound healing. In the present work, the structural and biological features of hAM are introduced as well as the application of this layer in different forms of composites to enhance wound healing. Future studies are recommended to detect possible further functionalization to enhance the hAM effectiveness on wound healing.
Collapse
Affiliation(s)
- Parisa Heydari
- Department of Biomaterials Nanotechnology and Tissue Engineering, School of Advanced Technology in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Mojahedi
- Department of Biomaterials Nanotechnology and Tissue Engineering, School of Advanced Technology in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Pouya Javaherchi
- Department of Biomaterials Nanotechnology and Tissue Engineering, School of Advanced Technology in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maede Sharifi
- Department of Biomaterials Nanotechnology and Tissue Engineering, School of Advanced Technology in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Anousheh Zargar Kharazi
- Department of Biomaterials Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Applied Physiology Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
14
|
Shi J, Yao H, Chong H, Hu X, Yang J, Dai X, Liu D, Wu Z, Dang M, Fei W, Wang DA. Tissue-engineered collagen matrix loaded with rat adipose-derived stem cells/human amniotic mesenchymal stem cells for rotator cuff tendon-bone repair. Int J Biol Macromol 2024; 282:137144. [PMID: 39488324 DOI: 10.1016/j.ijbiomac.2024.137144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/05/2024] [Accepted: 10/30/2024] [Indexed: 11/04/2024]
Abstract
The rotator cuff tendon-bone interface tissue exhibits high heterogeneity in its composition and structure, with collagen being its primary component. Here, we prepared tissue-engineered decellularized live hyaline cartilage grafts (dLHCG), this dLHCG scaffold's bioactive ECM mainly consists of collagen II, proteoglycans, and fibronectin, presenting a cartilage-like lacuna microstructure. The dLHCG scaffold loaded human amniotic mesenchymal stem cells (hAMSCs) and adipose stem cells (ADSCs) were implanted into the interface. The dLHCG scaffold could maintain the pluripotency of stem cells, supporting the proliferation, osteogenic differentiation, and tenogenic differentiation of the MSCs. The collagen II, through the integrin α2β1-FAK-JNK signaling axis, promotes Runx-2 activation, playing a better regulatory role in the early osteogenic differentiation of MSCs, enhancing bone defect repair through an endochondral ossification process. The in vivo rat model demonstrated that 12 weeks post-operation, the MSC-loaded dLHCG scaffold group exhibited continuous aligned collagen fibers at the tendon-bone interface, with significantly enhanced biomechanical function compared to the control group. The dLHCG scaffold create an efficient interface, which promoting the restoration of the soft-hard gradient structure tissue at the junction between the scaffold and the host tissue, thereby providing a rational and promising strategy for the rapid healing of the rotator cuff injury.
Collapse
Affiliation(s)
- Junli Shi
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, PR China
| | - Hang Yao
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, PR China.
| | - Hui Chong
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, PR China
| | - Xu Hu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
| | - Jian Yang
- Clinical Medical College, Yangzhou University, Yangzhou 225001, PR China
| | - Xiaomei Dai
- Department of Orthopedics and Sports medicine, Northern Jiangsu People's Hospital, Yangzhou 225001, PR China
| | - Dianwei Liu
- Dalian Medical University, Dalian 116044, PR China
| | - Zhonglian Wu
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, PR China
| | - Mengbo Dang
- Dalian Medical University, Dalian 116044, PR China
| | - Wenyong Fei
- Department of Orthopedics and Sports medicine, Northern Jiangsu People's Hospital, Yangzhou 225001, PR China; Center of Basic and Clinical Research in Sports Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, PR China.
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong; Shenzhen Research Institute, City University of Hong Kong, Shenzhen, PR China.
| |
Collapse
|
15
|
Rodríguez-Valiente M, García-Hernández AM, Fuente-Mora C, Sánchez-Gálvez J, García-Vizcaino EM, Tristante Barrenechea E, Castellanos Escrig G, Liarte Lastra SD, Nicolás FJ. Management of Foot Ulcers and Chronic Wounds with Amniotic Membrane in Comorbid Patients: A Successful Experience. Biomedicines 2024; 12:2380. [PMID: 39457692 PMCID: PMC11505069 DOI: 10.3390/biomedicines12102380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Chronic wounds are defined as those with disturbances in normal healing. They involve symptoms like exudate, odor, pain or impaired mobility, severely impacting life quality. In the case of patients with additional comorbidities, these are known to aggravate the healing impairment. Amniotic membrane (AM) is gaining attention for its regenerative potential, as it has shown promise in treating hard-to-heal wounds, such as diabetic foot ulcers. This work examines a series of five patients who, while suffering an array of other chronic conditions, were treated with AM for the management of non-healing chronic ulcers. Inclusion criteria involved patients with lesions that have been active at least for six weeks and resistant to multiple treatments, accompanied by complex underlying pathologies affecting cardiovascular, immune or renal functions. Exclusion criteria included untreated active infections and patients undergoing other experimental treatments. The mean age of the patients was 68.4 ± 5.2 years. Wounds were treated once a week with AM, following standardized procedures. The variables measured included pain levels, microorganism presence, wound reduction and the number of AM applications to recovery. The median pain VAS score decreased significantly from seven at the start to two at the end of procedures. Four out of five patients achieved complete epithelialization, while the remaining patient showed significant reductions of 40% in wound size after 14 months. Our results confirm how the application of AM is a safe and effective resource for the management of chronic wounds in patients with serious comorbidities, enhancing patients' quality of life, firstly by reducing pain, later by allowing recovery. Future research, including molecular analyses of wound exudates before and after AM treatment, can contribute to better understanding and fine tuning of this therapeutic resource.
Collapse
Affiliation(s)
- Mónica Rodríguez-Valiente
- Department of General Surgery, Hospital Clínico Universitario Virgen de la Arrixaca, Biomedical Research Institute of Murcia (IMIB-Arrixaca), El Palmar, 30120 Murcia, Spain;
- Faculty of Nursing, UCAM Catholic University of Murcia, Av. Jerónimos 135, Guadalupe, 30107 Murcia, Spain; (J.S.-G.); (E.M.G.-V.)
- Research Group on Molecular and Cellular Biology Solutions in Regenerative Medicine, UCAM Catholic University of Murcia, Av. Jerónimos 135, Guadalupe, 30107 Murcia, Spain
| | - Ana M. García-Hernández
- Cell Therapy Unit at Hospital Clínico Universitario Virgen de la Arrixaca, Biomedical Research Institute of Murcia (IMIB-Arrixaca), El Palmar, 30120 Murcia, Spain
| | - Cristina Fuente-Mora
- Clinical Trial Plataforms FFIS, Biomedical Research Institute of Murcia (IMIB-Arrixaca), El Palmar, 30120 Murcia, Spain;
| | - Javier Sánchez-Gálvez
- Faculty of Nursing, UCAM Catholic University of Murcia, Av. Jerónimos 135, Guadalupe, 30107 Murcia, Spain; (J.S.-G.); (E.M.G.-V.)
- Research Group on Molecular and Cellular Biology Solutions in Regenerative Medicine, UCAM Catholic University of Murcia, Av. Jerónimos 135, Guadalupe, 30107 Murcia, Spain
| | - Eva María García-Vizcaino
- Faculty of Nursing, UCAM Catholic University of Murcia, Av. Jerónimos 135, Guadalupe, 30107 Murcia, Spain; (J.S.-G.); (E.M.G.-V.)
- Research Group on Molecular and Cellular Biology Solutions in Regenerative Medicine, UCAM Catholic University of Murcia, Av. Jerónimos 135, Guadalupe, 30107 Murcia, Spain
| | - Elena Tristante Barrenechea
- Cell Therapy Unit at Hospital Clínico Universitario Virgen de la Arrixaca, Biomedical Research Institute of Murcia (IMIB-Arrixaca), El Palmar, 30120 Murcia, Spain
| | - Gregorio Castellanos Escrig
- Department of General Surgery, Hospital Clínico Universitario Virgen de la Arrixaca, Biomedical Research Institute of Murcia (IMIB-Arrixaca), El Palmar, 30120 Murcia, Spain;
| | - Sergio David Liarte Lastra
- Faculty of Nursing, UCAM Catholic University of Murcia, Av. Jerónimos 135, Guadalupe, 30107 Murcia, Spain; (J.S.-G.); (E.M.G.-V.)
- Research Group on Molecular and Cellular Biology Solutions in Regenerative Medicine, UCAM Catholic University of Murcia, Av. Jerónimos 135, Guadalupe, 30107 Murcia, Spain
| | - Francisco Jose Nicolás
- Laboratorio de Regeneración, Oncología Molecular y TGF-ß IMIB-Arrixaca, Biomedical Research Institute of Murcia (IMIB-Arrixaca), El Palmar, 30120 Murcia, Spain;
| |
Collapse
|
16
|
Moghimi MH, Salehian M, Abdi M, Tahrekhani M, Safaei A, Kamali K. The impact of an open-label design on human amniotic membranes vs. silver sulfadiazine dressings for second-degree burns: a randomized controlled clinical trial. BMC Surg 2024; 24:309. [PMID: 39396946 PMCID: PMC11472429 DOI: 10.1186/s12893-024-02554-5] [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: 10/21/2023] [Accepted: 09/02/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND Burn wounds require optimal medical management due to associated psycho-emotional and socioeconomic impacts and severe pain. The use of synthetic and biological dressings improves healing and reduces burn wound complications. The present study aimed to compare the outcomes of using human amniotic membrane (hAM) dressings and conventional silver sulfadiazine (SSDZ) ointment dressings in the management of second-degree burn wounds. METHODS Fifty patients who participated in this clinical trial were divided into two groups via simple randomization. All the enrolled patients, who had burnt in the last 24 h, had thermal damage mechanisms and were suffering from less than 20% second-degree heat-burn wounds on the skin surface. The target group (n = 25) was treated with hAM, and the control group (n = 25) was treated with SSDZ ointment. The researcher-designed checklist was used to determine the clinical performance in the follow-up assessments on days 7, 14, and 30. RESULTS No significant differences were detected in terms of sex, age, or percentage of burn wounds (p > 0.05). Wound epithelialization at days 7, 14, and 30, scar formation, wound pigmentation, pain severity, analgesia requirements, and hospital stay length (on day 30) were significantly lower in the target group (treated with hAM) than in the control group (treated with SSDZ ointment) (p < 0.05). However, treatment costs in the target group ($170) were significantly higher than those in the control group ($71) (p < 0.001). CONCLUSION Despite its higher cost, hAM, as a technology-based therapy dressing, demonstrates superiority over SSDZ ointment in terms of wound healing and pain management.
Collapse
Affiliation(s)
- Mohammad Hossein Moghimi
- Department of General Surgery, School of Medicine, Ayatollah Mousavi Hospital, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehran Salehian
- Departmentbof Surgery, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Mohammad Abdi
- Department of Emergency and Critical Care, School of Nursing, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehran Tahrekhani
- Department of Medical-Surgical, Abhar School of Nursing, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Alireza Safaei
- School of Science, Engineering, and Environment, University of Salford, Manchester, UK
| | - Koorosh Kamali
- Department of Epidemiology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
17
|
Perry AG, Kahn A, Mercuri J, Rini K, Chang J, Pathak RA. Preclinical and clinical evidence for using perinatal tissue allografts in nerve sparing robot assisted radical prostatectomy to hasten recovery of functional outcomes: a literature review. BMC Urol 2024; 24:208. [PMID: 39342266 PMCID: PMC11438271 DOI: 10.1186/s12894-024-01593-7] [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] [Accepted: 09/12/2024] [Indexed: 10/01/2024] Open
Abstract
INTRODUCTION Localized prostate cancer (PCa) is one of the most common malignancies in the United States. Despite continued refinement of robot assisted radical prostatectomy (RARP) surgical methods, post-surgical erectile dysfunction and urinary incontinence remain significant challenges due to iatrogenic injury of local nervous tissue. Thus, the development of therapeutic strategies, including the use of biologic adjuncts to protect and/or enhance recovery and function of nerves following RARP is of growing interest. Perinatal tissue allografts have been investigated as one such biologic adjunct to nerve sparing RARP. However, knowledge regarding their clinical efficacy in hastening return of potency and continence as well as the potential underpinning biological mechanisms involved remains understudied. Thus, the objective of this literature review was to summarize published basic science and clinical studies supporting and evaluating the use of perinatal allografts for nerve repair and their clinical efficacy as adjuncts to RARP, respectively. METHODS The literature as of May 2024 was reviewed non-systematically using PubMed, EMBASE, Scopus, and Web of Science databases. The search terms utilized were "robotic prostatectomy", "prostate cancer", "nerve sparing", "perinatal tissue", "allograft", "potency", and "continence" alone or in combination. All articles were reviewed and judged for scientific merit by authors RP and JM, only peer-reviewed studies were considered. RESULTS Eight studies of perinatal tissue allograph use in RARP were deemed worthy of inclusion in this nonsystematic review. CONCLUSIONS Incontinence and impotence remain significant comorbidities despite continued advancement in surgical technique. However, basic science research has demonstrated potential neurotrophic, anti-fibrotic, and anti-inflammatory properties of perinatal tissue allografts, and clinical studies have shown that patients who receive an intra-operative prostatic perinatal membrane wrap have faster return to potency and continence.
Collapse
Affiliation(s)
- Alan G Perry
- Department of Urology, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL, 32224, USA
| | - Amanda Kahn
- Department of Urology, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL, 32224, USA
| | - Jeremy Mercuri
- Samaritan Biologics, Cordova, TN, USA
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | | | | | - Ram A Pathak
- Department of Urology, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL, 32224, USA.
| |
Collapse
|
18
|
Ingraldi AL, Allen T, Tinghitella JN, Merritt WC, Becker T, Tabor AJ. Characterization of Amnion-Derived Membrane for Clinical Wound Applications. Bioengineering (Basel) 2024; 11:953. [PMID: 39451330 PMCID: PMC11504399 DOI: 10.3390/bioengineering11100953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Human amniotic membrane (hAM), the innermost placental layer, has unique properties that allow for a multitude of clinical applications. It is a common misconception that birth-derived tissue products, such as dual-layered dehydrated amnion-amnion graft (dHAAM), are similar regardless of the manufacturing steps. A commercial dHAAM product, Axolotl Biologix DualGraft™, was assessed for biological and mechanical characteristics. Testing of dHAAM included antimicrobial, cellular biocompatibility, proteomics analysis, suture strength, and tensile, shear, and compressive modulus testing. Results demonstrated that the membrane can be a scaffold for fibroblast growth (cellular biocompatibility), containing an average total of 7678 unique proteins, 82,296 peptides, and 96,808 peptide ion variants that may be antimicrobial. Suture strength results showed an average pull force of 0.2 N per dHAAM sample (equating to a pull strength of 8.5 MPa). Tensile modulus data revealed variation, with wet samples showing 5× lower stiffness than dry samples. The compressive modulus and shear modulus displayed differences between donors (lots). This study emphasizes the need for standardized processing protocols to ensure consistency across dHAAM products and future research to explore comparative analysis with other amniotic membrane products. These findings provide baseline data supporting the potential of amniotic membranes in clinical applications.
Collapse
Affiliation(s)
| | - Tim Allen
- Axolotl Biologix, Scottsdale, AZ 85260, USA; (A.L.I.)
| | | | - William C. Merritt
- Mechanical Engineering and Center for Materials Interfaces in Research and Applications (MIRA), Northern Arizona University, Flagstaff, AZ 86011, USA; (W.C.M.)
| | - Timothy Becker
- Mechanical Engineering and Center for Materials Interfaces in Research and Applications (MIRA), Northern Arizona University, Flagstaff, AZ 86011, USA; (W.C.M.)
| | - Aaron J. Tabor
- Axolotl Biologix, Scottsdale, AZ 85260, USA; (A.L.I.)
- Biological Sciences, Northern Arizona University, Flagstaff, AZ 86011, USA;
| |
Collapse
|
19
|
Marassi V, La Rocca G, Placci A, Muntiu A, Vincenzoni F, Vitali A, Desiderio C, Maraldi T, Beretti F, Russo E, Miceli V, Conaldi PG, Papait A, Romele P, Cargnoni A, Silini AR, Alviano F, Parolini O, Giordani S, Zattoni A, Reschiglian P, Roda B. Native characterization and QC profiling of human amniotic mesenchymal stromal cell vesicular fractions for secretome-based therapy. Talanta 2024; 276:126216. [PMID: 38761653 DOI: 10.1016/j.talanta.2024.126216] [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: 12/18/2023] [Revised: 04/09/2024] [Accepted: 05/05/2024] [Indexed: 05/20/2024]
Abstract
Human amniotic mesenchymal stromal cells (hAMSCs) have unique immunomodulatory properties making them attractive candidates for regenerative applications in inflammatory diseases. Most of their beneficial properties are mediated through their secretome. The bioactive factors concurring to its therapeutic activity are still unknown. Evidence suggests synergy between the two main components of the secretome, soluble factors and vesicular fractions, pivotal in shifting inflammation and promoting self-healing. Biological variability and the absence of quality control (QC) protocols hinder secretome-based therapy translation to clinical applications. Moreover, vesicular secretome contains a multitude of particles with varying size, cargos and functions whose complexity hinders full characterization and comprehension. This study achieved a significant advancement in secretome characterization by utilizing native, FFF-based separation and characterizing extracellular vesicles derived from hAMSCs. This was accomplished by obtaining dimensionally homogeneous fractions then characterized based on their protein content, potentially enabling the identification of subpopulations with diverse functionalities. This method proved to be successful as an independent technique for secretome profiling, with the potential to contribute to the standardization of a qualitative method. Additionally, it served as a preparative separation tool, streamlining populations before ELISA and LC-MS characterization. This approach facilitated the categorization of distinctive and recurring proteins, along with the identification of clusters associated with vesicle activity and functions. However, the presence of proteins unique to each fraction obtained through the FFF separation tool presents a challenge for further analysis of the protein content within these cargoes.
Collapse
Affiliation(s)
- Valentina Marassi
- Department of Chemistry G. Ciamician, University of Bologna, Italy; byFlow srl, Bologna, Italy
| | - Giampiero La Rocca
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127, Palermo, Italy
| | - Anna Placci
- Department of Chemistry G. Ciamician, University of Bologna, Italy
| | - Alexandra Muntiu
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", Consiglio Nazionale delle Ricerche, 00168, Rome, Italy
| | - Federica Vincenzoni
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 00168, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Alberto Vitali
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", Consiglio Nazionale delle Ricerche, 00168, Rome, Italy
| | - Claudia Desiderio
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", Consiglio Nazionale delle Ricerche, 00168, Rome, Italy
| | - Tullia Maraldi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Francesca Beretti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Eleonora Russo
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127, Palermo, Italy
| | - Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), 90127, Palermo, Italy
| | - Pier Giulio Conaldi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), 90127, Palermo, Italy
| | - Andrea Papait
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy; Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Pietro Romele
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124, Brescia, Italy
| | - Anna Cargnoni
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124, Brescia, Italy
| | - Antonietta Rosa Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124, Brescia, Italy
| | - Francesco Alviano
- Department of Biomedical and Neuromotor Science, University of Bologna, Bologna, Italy
| | - Ornella Parolini
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy; Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Stefano Giordani
- Department of Chemistry G. Ciamician, University of Bologna, Italy
| | - Andrea Zattoni
- Department of Chemistry G. Ciamician, University of Bologna, Italy; byFlow srl, Bologna, Italy
| | - Pierluigi Reschiglian
- Department of Chemistry G. Ciamician, University of Bologna, Italy; byFlow srl, Bologna, Italy
| | - Barbara Roda
- Department of Chemistry G. Ciamician, University of Bologna, Italy; byFlow srl, Bologna, Italy.
| |
Collapse
|
20
|
Masserdotti A, Gasik M, Grillari-Voglauer R, Grillari J, Cargnoni A, Chiodelli P, Papait A, Magatti M, Romoli J, Ficai S, Di Pietro L, Lattanzi W, Silini AR, Parolini O. Unveiling the human fetal-maternal interface during the first trimester: biophysical knowledge and gaps. Front Cell Dev Biol 2024; 12:1411582. [PMID: 39144254 PMCID: PMC11322133 DOI: 10.3389/fcell.2024.1411582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/11/2024] [Indexed: 08/16/2024] Open
Abstract
The intricate interplay between the developing placenta and fetal-maternal interactions is critical for pregnancy outcomes. Despite advancements, gaps persist in understanding biomechanics, transport processes, and blood circulation parameters, all of which are crucial for safe pregnancies. Moreover, the complexity of fetal-maternal interactions led to conflicting data and methodological variations. This review presents a comprehensive overview of current knowledge on fetal-maternal interface structures, with a particular focus on the first trimester. More in detail, the embryological development, structural characteristics, and physiological functions of placental chorionic plate and villi, fetal membranes and umbilical cord are discussed. Furthermore, a description of the main structures and features of maternal and fetal fluid dynamic exchanges is provided. However, ethical constraints and technological limitations pose still challenges to studying early placental development directly, which calls for sophisticated in vitro, microfluidic organotypic models for advancing our understanding. For this, knowledge about key in vivo parameters are necessary for their design. In this scenario, the integration of data from later gestational stages and mathematical/computational simulations have proven to be useful tools. Notwithstanding, further research into cellular and molecular mechanisms at the fetal-maternal interface is essential for enhancing prenatal care and improving maternal and fetal health outcomes.
Collapse
Affiliation(s)
- Alice Masserdotti
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | | | - Johannes Grillari
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
- Institute of Molecular Biotechnology, BOKU University, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Austria
| | - Anna Cargnoni
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Paola Chiodelli
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Papait
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| | - Marta Magatti
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Jacopo Romoli
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Sara Ficai
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lorena Di Pietro
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| | - Wanda Lattanzi
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| | - Antonietta Rosa Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| |
Collapse
|
21
|
Goushki MA, Kharat Z, Kehtari M, Sohi AN, Ahvaz HH, Rad I, HosseinZadeh S, Kouhkan F, Kabiri M. Applications of extraembryonic tissue-derived cells in vascular tissue regeneration. Stem Cell Res Ther 2024; 15:205. [PMID: 38982541 PMCID: PMC11234723 DOI: 10.1186/s13287-024-03784-3] [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: 02/29/2024] [Accepted: 06/06/2024] [Indexed: 07/11/2024] Open
Abstract
Vascular tissue engineering is a promising approach for regenerating damaged blood vessels and developing new therapeutic approaches for heart disease treatment. To date, different sources of cells have been recognized that offer assistance within the recovery of heart supply routes and veins with distinctive capacities and are compelling for heart regeneration. However, some challenges still remain that need to be overcome to establish the full potential application of these cells. In this paper, we review the different cell sources used for vascular tissue engineering, focusing on extraembryonic tissue-derived cells (ESCs), and elucidate their roles in cardiovascular disease. In addition, we highlight the intricate interplay between mechanical and biochemical factors in regulating mesenchymal stem cell (MSC) differentiation, offering insights into optimizing their application in vascular tissues.
Collapse
Affiliation(s)
- Mehdi Amiri Goushki
- Department of Life Science Engineering, Faculty of New Sciences & Technologies, University of Tehran, Tehran, 14395-1561, Iran
| | - Zahra Kharat
- Department of Life Science Engineering, Faculty of New Sciences & Technologies, University of Tehran, Tehran, 14395-1561, Iran
| | - Mousa Kehtari
- School of Biology, College of Sciences, University of Tehran, Tehran, 1417614411, Iran
| | - Alireza Naderi Sohi
- National Institute of Genetic Engineering and Biotechnology, Tehran, 1497716316, Iran
| | | | - Iman Rad
- Stem Cell Technology Research Center, Tehran, 15856-36473, Iran
| | - Simzar HosseinZadeh
- Department of Tissue Engineering and Regenerative Medicine, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Kouhkan
- Stem Cell Technology Research Center, Tehran, 15856-36473, Iran
| | - Mahboubeh Kabiri
- Department of Biotechnology, College of Science, University of Tehran, Tehran, 14155-6455, Iran.
| |
Collapse
|
22
|
Mello DB, Mesquita FCP, Silva dos Santos D, Asensi KD, Dias ML, Campos de Carvalho AC, Goldenberg RCDS, Kasai-Brunswick TH. Mesenchymal Stromal Cell-Based Products: Challenges and Clinical Therapeutic Options. Int J Mol Sci 2024; 25:6063. [PMID: 38892249 PMCID: PMC11173248 DOI: 10.3390/ijms25116063] [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: 05/02/2024] [Revised: 05/24/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
Mesenchymal stromal cell (MSC)-based advanced therapy medicinal products (ATMPs) are being tried in a vast range of clinical applications. These cells can be isolated from different donor tissues by using several methods, or they can even be derived from induced pluripotent stem cells or embryonic stem cells. However, ATMP heterogeneity may impact product identity and potency, and, consequently, clinical trial outcomes. In this review, we discuss these topics and the need to establish minimal criteria regarding the manufacturing of MSCs so that these innovative therapeutics may be better positioned to contribute to the advancement of regenerative medicine.
Collapse
Affiliation(s)
- Debora B. Mello
- National Center of Structural Biology and Bioimaging, CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.B.M.); (A.C.C.d.C.)
| | | | - Danúbia Silva dos Santos
- Center of Cellular Technology, National Institute of Cardiology, INC, Rio de Janeiro 22240-002, Brazil;
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
| | - Karina Dutra Asensi
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Marlon Lemos Dias
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Antonio Carlos Campos de Carvalho
- National Center of Structural Biology and Bioimaging, CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.B.M.); (A.C.C.d.C.)
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Regina Coeli dos Santos Goldenberg
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Tais Hanae Kasai-Brunswick
- National Center of Structural Biology and Bioimaging, CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.B.M.); (A.C.C.d.C.)
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| |
Collapse
|
23
|
Calligaris M, Zito G, Busà R, Bulati M, Iannolo G, Gallo A, Carreca AP, Cuscino N, Castelbuono S, Carcione C, Centi C, Amico G, Bertani A, Chinnici CM, Conaldi PG, Scilabra SD, Miceli V. Proteomic analysis and functional validation reveal distinct therapeutic capabilities related to priming of mesenchymal stromal/stem cells with IFN-γ and hypoxia: potential implications for their clinical use. Front Cell Dev Biol 2024; 12:1385712. [PMID: 38882056 PMCID: PMC11179434 DOI: 10.3389/fcell.2024.1385712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 05/13/2024] [Indexed: 06/18/2024] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) are a heterogeneous population of multipotent cells that can be obtained from various tissues, such as dental pulp, adipose tissue, bone marrow and placenta. MSCs have gained importance in the field of regenerative medicine because of their promising role in cell therapy and their regulatory abilities in tissue repair and regeneration. However, a better characterization of these cells and their products is necessary to further potentiate their clinical application. In this study, we used unbiased high-resolution mass spectrometry-based proteomic analysis to investigate the impact of distinct priming strategies, such as hypoxia and IFN-γ treatment, on the composition and therapeutic functionality of the secretome produced by MSCs derived from the amniotic membrane of the human placenta (hAMSCs). Our investigation revealed that both types of priming improved the therapeutic efficacy of hAMSCs, and these improvements were related to the secretion of functional factors present in the conditioned medium (CM) and exosomes (EXOs), which play crucial roles in mediating the paracrine effects of MSCs. In particular, hypoxia was able to induce a pro-angiogenic, innate immune response-activating, and tissue-regenerative hAMSC phenotype, as highlighted by the elevated production of regulatory factors such as VEGFA, PDGFRB, ANGPTL4, ENG, GRO-γ, IL8, and GRO-α. IFN-γ priming, instead, led to an immunosuppressive profile in hAMSCs, as indicated by increased levels of TGFB1, ANXA1, THBS1, HOMER2, GRN, TOLLIP and MCP-1. Functional assays validated the increased angiogenic properties of hypoxic hAMSCs and the enhanced immunosuppressive activity of IFN-γ-treated hAMSCs. This study extends beyond the direct priming effects on hAMSCs, demonstrating that hypoxia and IFN-γ can influence the functional characteristics of hAMSC-derived secretomes, which, in turn, orchestrate the production of functional factors by peripheral blood cells. This research provides valuable insights into the optimization of MSC-based therapies by systematically assessing and comparing the priming type-specific functional features of hAMSCs. These findings highlight new strategies for enhancing the therapeutic efficacy of MSCs, particularly in the context of multifactorial diseases, paving the way for the use of hAMSC-derived products in clinical practice.
Collapse
Affiliation(s)
- Matteo Calligaris
- Proteomics Group, Ri.MED Foundation c/o IRCCS ISMETT, Palermo, Italy
| | - Giovanni Zito
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo, Italy
| | - Rosalia Busà
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo, Italy
| | - Matteo Bulati
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo, Italy
| | - Gioacchin Iannolo
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo, Italy
| | - Alessia Gallo
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo, Italy
| | | | - Nicola Cuscino
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo, Italy
| | - Salvatore Castelbuono
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo, Italy
| | | | - Claudio Centi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo, Italy
| | | | - Alessandro Bertani
- Thoracic Surgery and Lung Transplantation Unit, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo, Italy
| | - Cinzia Maria Chinnici
- Regenerative Medicine and Immunotherapy Area, Ri.MED Foundation c/o IRCCS ISMETT, Palermo, Italy
| | - Pier Giulio Conaldi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo, Italy
| | | | - Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo, Italy
| |
Collapse
|
24
|
Arellano MYG, VanHeest M, Emmadi S, Abdul-Hafez A, Ibrahim SA, Thiruvenkataramani RP, Teleb RS, Omar H, Kesaraju T, Mohamed T, Madhukar BV, Omar SA. Role of Mesenchymal Stem/Stromal Cells (MSCs) and MSC-Derived Extracellular Vesicles (EVs) in Prevention of Telomere Length Shortening, Cellular Senescence, and Accelerated Biological Aging. Bioengineering (Basel) 2024; 11:524. [PMID: 38927760 PMCID: PMC11200821 DOI: 10.3390/bioengineering11060524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 06/28/2024] Open
Abstract
Biological aging is defined as a progressive decline in tissue function that eventually results in cell death. Accelerated biologic aging results when the telomere length is shortened prematurely secondary to damage from biological or environmental stressors, leading to a defective reparative mechanism. Stem cells therapy may have a potential role in influencing (counteract/ameliorate) biological aging and maintaining the function of the organism. Mesenchymal stem cells, also called mesenchymal stromal cells (MSCs) are multipotent stem cells of mesodermal origin that can differentiate into other types of cells, such as adipocytes, chondrocytes, and osteocytes. MSCs influence resident cells through the secretion of paracrine bioactive components such as cytokines and extracellular vesicles (EVs). This review examines the changes in telomere length, cellular senescence, and normal biological age, as well as the factors contributing to telomere shortening and accelerated biological aging. The role of MSCs-especially those derived from gestational tissues-in prevention of telomere shortening (TS) and accelerated biological aging is explored. In addition, the strategies to prevent MSC senescence and improve the antiaging therapeutic application of MSCs and MSC-derived EVs in influencing telomere length and cellular senescence are reviewed.
Collapse
Affiliation(s)
- Myrna Y. Gonzalez Arellano
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Regional Neonatal Intensive Care Unit, Sparrow Hospital, Lansing, MI 48912, USA
| | - Matthew VanHeest
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
| | - Sravya Emmadi
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
| | - Amal Abdul-Hafez
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
| | - Sherif Abdelfattah Ibrahim
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Histology and Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Ranga P. Thiruvenkataramani
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Regional Neonatal Intensive Care Unit, Sparrow Hospital, Lansing, MI 48912, USA
| | - Rasha S. Teleb
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Department of Pediatrics and Neonatology, Qena Faculty of Medicine, South Valley University, Qena 83523, Egypt
| | - Hady Omar
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
| | - Tulasi Kesaraju
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
| | - Tarek Mohamed
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Regional Neonatal Intensive Care Unit, Sparrow Hospital, Lansing, MI 48912, USA
| | - Burra V. Madhukar
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
| | - Said A. Omar
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Regional Neonatal Intensive Care Unit, Sparrow Hospital, Lansing, MI 48912, USA
| |
Collapse
|
25
|
Sulcanese L, Prencipe G, Canciello A, Cerveró-Varona A, Perugini M, Mauro A, Russo V, Barboni B. Stem-Cell-Driven Chondrogenesis: Perspectives on Amnion-Derived Cells. Cells 2024; 13:744. [PMID: 38727280 PMCID: PMC11083072 DOI: 10.3390/cells13090744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Regenerative medicine harnesses stem cells' capacity to restore damaged tissues and organs. In vitro methods employing specific bioactive molecules, such as growth factors, bio-inductive scaffolds, 3D cultures, co-cultures, and mechanical stimuli, steer stem cells toward the desired differentiation pathways, mimicking their natural development. Chondrogenesis presents a challenge for regenerative medicine. This intricate process involves precise modulation of chondro-related transcription factors and pathways, critical for generating cartilage. Cartilage damage disrupts this process, impeding proper tissue healing due to its unique mechanical and anatomical characteristics. Consequently, the resultant tissue often forms fibrocartilage, which lacks adequate mechanical properties, posing a significant hurdle for effective regeneration. This review comprehensively explores studies showcasing the potential of amniotic mesenchymal stem cells (AMSCs) and amniotic epithelial cells (AECs) in chondrogenic differentiation. These cells exhibit innate characteristics that position them as promising candidates for regenerative medicine. Their capacity to differentiate toward chondrocytes offers a pathway for developing effective regenerative protocols. Understanding and leveraging the innate properties of AMSCs and AECs hold promise in addressing the challenges associated with cartilage repair, potentially offering superior outcomes in tissue regeneration.
Collapse
Affiliation(s)
- Ludovica Sulcanese
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Angelo Canciello
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Monia Perugini
- Department of Bioscience and Technology for Food, Agriculture, and Environment, University of Teramo, 64100 Teramo, Italy;
| | - Annunziata Mauro
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Valentina Russo
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| |
Collapse
|
26
|
Lv Y, Yu W, Xuan R, Yang Y, Xue X, Ma X. Human Placental Mesenchymal Stem Cells-Exosomes Alleviate Endothelial Barrier Dysfunction via Cytoskeletal Remodeling through hsa-miR-148a-3p/ROCK1 Pathway. Stem Cells Int 2024; 2024:2172632. [PMID: 38681858 PMCID: PMC11055650 DOI: 10.1155/2024/2172632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/18/2024] [Accepted: 04/02/2024] [Indexed: 05/01/2024] Open
Abstract
Background Endothelial barrier disruption of human pulmonary vascular endothelial cells (HPVECs) is an important pathogenic factor for acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). Mesenchymal stem cells-exosome (MSCs-Exo) represents an ideal carrier for cell-free therapy. The therapeutic implication and underlying mechanism of human placental MSCs-Exo (HPMSCs-Exo) in ALI/ARDS need to be further explored. Materials and Methods HPMSCs-Exo was extracted from HPMSCs and characterized. Then, the therapeutic effects of exosomes were evaluated in ALI mice and HPVECs. RNA-sequencing was applied to reveal the miRNA profile of HPMSCs-Exo and differentially expressed genes (DEGs) in HPMSCs-Exo-pretreated HPVECs. The targets of miRNAs were predicted by bioinformatics methods and correlated to DEGs. Finally, the role of hsa-miR-148a-3p/ROCK1 pathway in HPVECs has been further discussed. Results The results showed that HPMSCs-Exo could downregulate Rho-associated coiled-coil-containing protein kinase 1 (ROCK1), upregulate the expression of zonula occludens-1 (ZO-1) and F-actin, promote HPVECs migration and tube formation, reduce cytoskeletal disorders and cell permeability, and thus improve ALI/ARDS. RNA-sequencing revealed the DEGs were mainly enriched in cell junction, angiogenesis, inflammation, and energy metabolism. HPMSCs-Exo contains multiple miRNAs which are associated with cytoskeletal function; the expression abundance of hsa-miR-148a-3p is the highest. Bioinformatic analysis identified ROCK1 as a target of hsa-miR-148a-3p. The overexpression of hsa-miR-148a-3p in HPMSCs-Exo promoted the migration and tube formation of HPVECs and reduced ROCK1 expression. However, the overexpression of ROCK1 on HPVECs reduced the therapeutic effect of HPMSCs-Exo. Conclusions HPMSCs-Exo represents a protective regimen against endothelial barrier disruption of HPVECs in ALI/ARDS, and the hsa-miR-148a-3p/ROCK1 pathway plays an important role in this therapeutics implication.
Collapse
Affiliation(s)
- Yuzhen Lv
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
- Ningxia Institute for Human Stem Cell Research, General Hospital of Ningxia Medical University, Yinchuan 750003, China
| | - Wenqin Yu
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
- Ningxia Institute for Human Stem Cell Research, General Hospital of Ningxia Medical University, Yinchuan 750003, China
| | - Ruiui Xuan
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
| | - Yulu Yang
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
| | - Xiaolan Xue
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
| | - Xiaowei Ma
- Intensive Care Unit, Cardiocerebral Vascular Disease Hospital, General Hospital of Ningxia Medical University, Yinchuan 750003, China
| |
Collapse
|
27
|
Shih HM, Chen YC, Yeh YT, Peng FS, Wu SC. Assessment of the feasibility of human amniotic membrane stem cell-derived cardiomyocytes in vitro. Heliyon 2024; 10:e28398. [PMID: 38560255 PMCID: PMC10979088 DOI: 10.1016/j.heliyon.2024.e28398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 03/18/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Myocardial infarction (MI) is a leading cause of death worldwide, resulting in extensive loss of cardiomyocytes and subsequent heart failure. Inducing cardiac differentiation of stem cells is a potential approach for myocardial regeneration therapy to improve post-MI prognosis. Mesenchymal stem cells (MSCs) have several advantages, including immune privilege and multipotent differentiation potential. This study aimed to explore the feasibility of chemically inducing human amniotic membrane MSCs (hAMSCs) to differentiate into cardiomyocytes in vitro. Human amniotic membrane (AM) samples were obtained from routine cesarean sections at Far Eastern Memorial Hospital. The isolated cells exhibited spindle-shaped morphology and expressed surface antigens CD73, CD90, CD105, and CD44, while lacking expression of CD19, CD11b, CD19, CD45, and HLA-DR. The SSEA-1, SSEA-3, and SSEA-4 markers were also positive, and the cells displayed the ability for tri-lineage differentiation into adipocytes, chondrocytes, and osteoblasts. The expression levels of MLC2v, Nkx2.5, and MyoD were analyzed using qPCR after applying various protocols for chemical induction, including BMP4, ActivinA, 5-azacytidine, CHIR99021, and IWP2 on hAMSCs. The group treated with 5 ng/ml BMP4, 10 ng/ml Activin A, 10 μM 5-azacytidine, 7.5 μM CHIR99021, and 5 μM IWP 2 expressed the highest levels of these genes. Furthermore, immunofluorescence staining demonstrated the expression of α-actinin and Troponin T in this group. In conclusion, this study demonstrated that hAMSCs can be chemically induced to differentiate into cardiomyocyte-like cells in vitro. However, to improve the functionality of the differentiated cells, further investigation of inductive protocols and regimens is needed.
Collapse
Affiliation(s)
- Hsiu-Man Shih
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Yi-Chen Chen
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Yen-Ting Yeh
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
- Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | | | - Shinn-Chih Wu
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
- Center for Biotechnology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
28
|
Shokati A, Naser Moghadasi A, Ghashghaei A, Sahraian MA, Chahardouli B, Mousavi SA, Ai J, Nikbakht M. Good manufacturing practices production of human placental derived mesenchymal stem cells for therapeutic applications: focus on multiple sclerosis. Mol Biol Rep 2024; 51:460. [PMID: 38551770 DOI: 10.1007/s11033-024-09372-1] [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: 12/05/2023] [Accepted: 02/21/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Among neurological diseases, multiple sclerosis (MS) affects mostly young adults and can cause long-term disability. While most medications with approval from regulatory agencies are very effective in treating MS disease, they are unable to repair the tissue damage found in the central nervous system (CNS). Consequently, Cell-based therapy particularly using mesenchymal stem/stromal cells (MSCs), holds promise for neuroprotection and tissue repair in MS treatment. Furthermore, placenta-derived MSCs (PLMSCs) have shown the potential to treat MS due to their abundance, noninvasive isolation from discarded tissues, no ethical problems, anti-inflammatory, and reparative properties. Accordingly, good manufacturing practices (GMPs) plays a crucial part in clinical SCs manufacturing. The purpose of our article is to discuss GMP-grade PLMSC protocols for treating MS as well as other clinical applications. METHODS AND RESULTS Placental tissue obtained of a healthy donor during the caesarean delivery and PLMSCs isolated by GMP standards. Flow cytometry was used to assess the expression of the CD markers CD34, CD105, CD90, and CD73 in the MSCs and the mesodermal differentiation ability was evaluated. Furthermore, Genetic evaluation of PLMSCs was done by G-banded karyotyping and revealed no chromosomal instability. In spite of the anatomical origin of the starting material, PLMSCs using this method of culture were maternal in origin. CONCLUSIONS We hope that our protocol for clinical manufacturing of PLMSCs according to GMP standards will assist researchers in isolating MSCs from placental tissue for clinical and pre-clinical applications.
Collapse
Affiliation(s)
- Ameneh Shokati
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abdorreza Naser Moghadasi
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | - Andisheh Ghashghaei
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Sahraian
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Bahram Chahardouli
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Asadollah Mousavi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohsen Nikbakht
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Research Institute for Oncology, Hematology and Cell Therapy, Shariati Hospital, Tehran University of Medical Sciences, Kargar Shomali Street, P. O. Box.: 1411713131, Tehran, Iran.
| |
Collapse
|
29
|
Jafari A, Mirzaei Y, Mer AH, Rezaei-Tavirani M, Jafari Z, Niknejad H. Comparison of the effects of preservation methods on structural, biological, and mechanical properties of the human amniotic membrane for medical applications. Cell Tissue Bank 2024; 25:305-323. [PMID: 37840108 DOI: 10.1007/s10561-023-10114-z] [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: 08/17/2023] [Accepted: 09/20/2023] [Indexed: 10/17/2023]
Abstract
Amniotic membrane (AM), the innermost layer of the placenta, is an exceptionally effective biomaterial with divers applications in clinical medicine. It possesses various biological functions, including scar reduction, anti-inflammatory properties, support for epithelialization, as well as anti-microbial, anti-fibrotic and angio-modulatory effects. Furthermore, its abundant availability, cost-effectiveness, and ethical acceptability make it a compelling biomaterial in the field of medicine. Given the potential unavailability of fresh tissue when needed, the preservation of AM is crucial to ensure a readily accessible and continuous supply for clinical use. However, preserving the properties of AM presents a significant challenge. Therefore, the establishment of standardized protocols for the collection and preservation of AM is vital to ensure optimal tissue quality and enhance patient safety. Various preservation methods, such as cryopreservation, lyophilization, and air-drying, have been employed over the years. However, identifying a preservation method that effectively safeguards AM properties remains an ongoing endeavor. This article aims to review and discuss different sterilization and preservation procedures for AM, as well as their impacts on its histological, physical, and biochemical characteristics.
Collapse
Affiliation(s)
- Ameneh Jafari
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yousef Mirzaei
- Department of Medical Biochemical Analysis, Cihan University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Ali Hussein Mer
- Department of Nusring, Mergasour Technical Institute, Erbil Polytechnic University, Erbil, Iraq
| | | | - Zahra Jafari
- 9th Dey Manzariye Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Gerli MFM, Calà G, Beesley MA, Sina B, Tullie L, Sun KY, Panariello F, Michielin F, Davidson JR, Russo FM, Jones BC, Lee DDH, Savvidis S, Xenakis T, Simcock IC, Straatman-Iwanowska AA, Hirst RA, David AL, O'Callaghan C, Olivo A, Eaton S, Loukogeorgakis SP, Cacchiarelli D, Deprest J, Li VSW, Giobbe GG, De Coppi P. Single-cell guided prenatal derivation of primary fetal epithelial organoids from human amniotic and tracheal fluids. Nat Med 2024; 30:875-887. [PMID: 38438734 PMCID: PMC10957479 DOI: 10.1038/s41591-024-02807-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/05/2024] [Indexed: 03/06/2024]
Abstract
Isolation of tissue-specific fetal stem cells and derivation of primary organoids is limited to samples obtained from termination of pregnancies, hampering prenatal investigation of fetal development and congenital diseases. Therefore, new patient-specific in vitro models are needed. To this aim, isolation and expansion of fetal stem cells during pregnancy, without the need for tissue samples or reprogramming, would be advantageous. Amniotic fluid (AF) is a source of cells from multiple developing organs. Using single-cell analysis, we characterized the cellular identities present in human AF. We identified and isolated viable epithelial stem/progenitor cells of fetal gastrointestinal, renal and pulmonary origin. Upon culture, these cells formed clonal epithelial organoids, manifesting small intestine, kidney tubule and lung identity. AF organoids exhibit transcriptomic, protein expression and functional features of their tissue of origin. With relevance for prenatal disease modeling, we derived lung organoids from AF and tracheal fluid cells of congenital diaphragmatic hernia fetuses, recapitulating some features of the disease. AF organoids are derived in a timeline compatible with prenatal intervention, potentially allowing investigation of therapeutic tools and regenerative medicine strategies personalized to the fetus at clinically relevant developmental stages.
Collapse
Affiliation(s)
- Mattia Francesco Maria Gerli
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, London, UK.
- Great Ormond Street Institute of Child Health, University College London, London, UK.
| | - Giuseppe Calà
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, London, UK
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Max Arran Beesley
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Beatrice Sina
- Great Ormond Street Institute of Child Health, University College London, London, UK
- Politecnico di Milano, Milan, Italy
| | - Lucinda Tullie
- Great Ormond Street Institute of Child Health, University College London, London, UK
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
| | - Kylin Yunyan Sun
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, London, UK
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Francesco Panariello
- Armenise/Harvard Laboratory of Integrative Genomics, Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Federica Michielin
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Joseph R Davidson
- Great Ormond Street Institute of Child Health, University College London, London, UK
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, UK
| | - Francesca Maria Russo
- Department of Development and Regeneration, Woman and Child and UZ Leuven Clinical Department of Obstetrics and Gynaecology, KU Leuven, Leuven, Belgium
| | - Brendan C Jones
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Dani Do Hyang Lee
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Savvas Savvidis
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Theodoros Xenakis
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Ian C Simcock
- Great Ormond Street Institute of Child Health, University College London, London, UK
- Department of Radiology, Great Ormond Street Hospital, London, UK
| | | | - Robert A Hirst
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Anna L David
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, UK
- Department of Development and Regeneration, Woman and Child and UZ Leuven Clinical Department of Obstetrics and Gynaecology, KU Leuven, Leuven, Belgium
| | | | - Alessandro Olivo
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Simon Eaton
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Stavros P Loukogeorgakis
- Great Ormond Street Institute of Child Health, University College London, London, UK
- Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Davide Cacchiarelli
- Armenise/Harvard Laboratory of Integrative Genomics, Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy
- Genomics and Experimental Medicine Program, Scuola Superiore Meridionale, Naples, Italy
| | - Jan Deprest
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, UK
- Department of Development and Regeneration, Woman and Child and UZ Leuven Clinical Department of Obstetrics and Gynaecology, KU Leuven, Leuven, Belgium
| | - Vivian S W Li
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
| | | | - Paolo De Coppi
- Great Ormond Street Institute of Child Health, University College London, London, UK.
- Department of Development and Regeneration, Woman and Child and UZ Leuven Clinical Department of Obstetrics and Gynaecology, KU Leuven, Leuven, Belgium.
- Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
- Medical and Surgical Department of the Fetus, Newborn and Infant, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy.
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK.
| |
Collapse
|
31
|
Kitawi R, Ledger S, Kelleher AD, Ahlenstiel CL. Advances in HIV Gene Therapy. Int J Mol Sci 2024; 25:2771. [PMID: 38474018 DOI: 10.3390/ijms25052771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Early gene therapy studies held great promise for the cure of heritable diseases, but the occurrence of various genotoxic events led to a pause in clinical trials and a more guarded approach to progress. Recent advances in genetic engineering technologies have reignited interest, leading to the approval of the first gene therapy product targeting genetic mutations in 2017. Gene therapy (GT) can be delivered either in vivo or ex vivo. An ex vivo approach to gene therapy is advantageous, as it allows for the characterization of the gene-modified cells and the selection of desired properties before patient administration. Autologous cells can also be used during this process which eliminates the possibility of immune rejection. This review highlights the various stages of ex vivo gene therapy, current research developments that have increased the efficiency and safety of this process, and a comprehensive summary of Human Immunodeficiency Virus (HIV) gene therapy studies, the majority of which have employed the ex vivo approach.
Collapse
Affiliation(s)
- Rose Kitawi
- Kirby Institute, University of New South Wales, Kensington, NSW 2052, Australia
| | - Scott Ledger
- Kirby Institute, University of New South Wales, Kensington, NSW 2052, Australia
| | - Anthony D Kelleher
- Kirby Institute, University of New South Wales, Kensington, NSW 2052, Australia
- St. Vincent's Hospital, Darlinghurst, NSW 2010, Australia
- UNSW RNA Institute, University of New South Wales, Kensington, NSW 2052, Australia
| | - Chantelle L Ahlenstiel
- Kirby Institute, University of New South Wales, Kensington, NSW 2052, Australia
- UNSW RNA Institute, University of New South Wales, Kensington, NSW 2052, Australia
| |
Collapse
|
32
|
Kafili G, Niknejad H, Tamjid E, Simchi A. Amnion-derived hydrogels as a versatile platform for regenerative therapy: from lab to market. Front Bioeng Biotechnol 2024; 12:1358977. [PMID: 38468689 PMCID: PMC10925797 DOI: 10.3389/fbioe.2024.1358977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/09/2024] [Indexed: 03/13/2024] Open
Abstract
In recent years, the amnion (AM) has emerged as a versatile tool for stimulating tissue regeneration and has been of immense interest for clinical applications. AM is an abundant and cost-effective tissue source that does not face strict ethical issues for biomedical applications. The outstanding biological attributes of AM, including side-dependent angiogenesis, low immunogenicity, anti-inflammatory, anti-fibrotic, and antibacterial properties facilitate its usage for tissue engineering and regenerative medicine. However, the clinical usage of thin AM sheets is accompanied by some limitations, such as handling without folding or tearing and the necessity for sutures to keep the material over the wound, which requires additional considerations. Therefore, processing the decellularized AM (dAM) tissue into a temperature-sensitive hydrogel has expanded its processability and applicability as an injectable hydrogel for minimally invasive therapies and a source of bioink for the fabrication of biomimetic tissue constructs by recapitulating desired biochemical cues or pre-defined architectural design. This article reviews the multi-functionality of dAM hydrogels for various biomedical applications, including skin repair, heart treatment, cartilage regeneration, endometrium regeneration, vascular graft, dental pulp regeneration, and cell culture/carrier platform. Not only recent and cutting-edge research is reviewed but also available commercial products are introduced and their main features and shortcomings are elaborated. Besides the great potential of AM-derived hydrogels for regenerative therapy, intensive interdisciplinary studies are still required to modify their mechanical and biological properties in order to broaden their therapeutic benefits and biomedical applications. Employing additive manufacturing techniques (e.g., bioprinting), nanotechnology approaches (e.g., inclusion of various bioactive nanoparticles), and biochemical alterations (e.g., modification of dAM matrix with photo-sensitive molecules) are of particular interest. This review article aims to discuss the current function of dAM hydrogels for the repair of target tissues and identifies innovative methods for broadening their potential applications for nanomedicine and healthcare.
Collapse
Affiliation(s)
- Golara Kafili
- Center for Nanoscience and Nanotechnology, Institute for Convergence Science and Technology, Sharif University of Technology, Tehran, Iran
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elnaz Tamjid
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abdolreza Simchi
- Center for Nanoscience and Nanotechnology, Institute for Convergence Science and Technology, Sharif University of Technology, Tehran, Iran
- Department of Materials Science and Engineering, Sharif University of Technology, Tehran, Iran
- Center for Bioscience and Technology, Institute for Convergence Science and Technology, Sharif University of Technology, Tehran, Iran
| |
Collapse
|
33
|
Ragni E, Piccolo S, Papait A, De Luca P, Taiana M, Grieco G, Silini AR, Parolini O, de Girolamo L. Stable Housekeeping Genes in Bone Marrow, Adipose Tissue, and Amniotic Membrane-Derived Mesenchymal Stromal Cells for Orthopedic Regenerative Medicine Approaches. Int J Mol Sci 2024; 25:1461. [PMID: 38338737 PMCID: PMC10855448 DOI: 10.3390/ijms25031461] [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: 12/22/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
The therapeutic effect of mesenchymal stromal cells (MSCs) has been described for a variety of disorders, including those affecting musculoskeletal tissues. In this context, the literature reports several data about the regenerative effectiveness of MSCs derived from bone marrow, adipose tissue, and an amniotic membrane (BMSCs, ASCs, and hAMSCs, respectively), either when expanded or when acting as clinical-grade biologic pillars of products used at the point of care. To date, there is no evidence about the superiority of one source over the others from a clinical perspective. Therefore, a reliable characterization of the tissue-specific MSC types is mandatory to identify the most effective treatment, especially when tailored to the target disease. Because molecular characterization is a crucial parameter for cell definition, the need for reliable normalizers as housekeeping genes (HKGs) is essential. In this report, the stability levels of five commonly used HKGs (ACTB, EF1A, GAPDH, RPLP0, and TBP) were sifted into BMSCs, ASCs, and hAMSCs. Adult and fetal/neonatal MSCs showed opposite HKG stability rankings. Moreover, by analyzing MSC types side-by-side, comparison-specific HKGs emerged. The effect of less performant HKG normalization was also demonstrated in genes coding for factors potentially involved in and predicting MSC therapeutic activity for osteoarthritis as a model musculoskeletal disorder, where the choice of the most appropriate normalizer had a higher impact on the donors rather than cell populations when compared side-by-side. In conclusion, this work confirms HKG source-specificity for MSCs and suggests the need for cell-type specific normalizers for cell source or condition-tailored gene expression studies.
Collapse
Affiliation(s)
- Enrico Ragni
- Laboratorio di Biotecnologie Applicate all’Ortopedia, IRCCS Istituto Ortopedico Galeazzi, Via Cristina Belgioioso 173, 20157 Milano, Italy; (S.P.); (P.D.L.); (M.T.); (G.G.); (L.d.G.)
| | - Simona Piccolo
- Laboratorio di Biotecnologie Applicate all’Ortopedia, IRCCS Istituto Ortopedico Galeazzi, Via Cristina Belgioioso 173, 20157 Milano, Italy; (S.P.); (P.D.L.); (M.T.); (G.G.); (L.d.G.)
| | - Andrea Papait
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (A.P.); (O.P.)
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Roma, Italy
| | - Paola De Luca
- Laboratorio di Biotecnologie Applicate all’Ortopedia, IRCCS Istituto Ortopedico Galeazzi, Via Cristina Belgioioso 173, 20157 Milano, Italy; (S.P.); (P.D.L.); (M.T.); (G.G.); (L.d.G.)
| | - Michela Taiana
- Laboratorio di Biotecnologie Applicate all’Ortopedia, IRCCS Istituto Ortopedico Galeazzi, Via Cristina Belgioioso 173, 20157 Milano, Italy; (S.P.); (P.D.L.); (M.T.); (G.G.); (L.d.G.)
| | - Giulio Grieco
- Laboratorio di Biotecnologie Applicate all’Ortopedia, IRCCS Istituto Ortopedico Galeazzi, Via Cristina Belgioioso 173, 20157 Milano, Italy; (S.P.); (P.D.L.); (M.T.); (G.G.); (L.d.G.)
| | - Antonietta Rosa Silini
- Centro di Ricerca “E. Menni”, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy;
| | - Ornella Parolini
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (A.P.); (O.P.)
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Roma, Italy
| | - Laura de Girolamo
- Laboratorio di Biotecnologie Applicate all’Ortopedia, IRCCS Istituto Ortopedico Galeazzi, Via Cristina Belgioioso 173, 20157 Milano, Italy; (S.P.); (P.D.L.); (M.T.); (G.G.); (L.d.G.)
| |
Collapse
|
34
|
Abou-Shanab AM, Gaser OA, Salah RA, El-Badri N. Application of the Human Amniotic Membrane as an Adjuvant Therapy for the Treatment of Hepatocellular Carcinoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1470:129-146. [PMID: 38036871 DOI: 10.1007/5584_2023_792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related morbidity and mortality worldwide. Current therapeutic approaches suffer significant side effects and lack of clear understanding of their molecular targets. Recent studies reported the anticancer effects, immunomodulatory properties, and antiangiogenic effects of the human amniotic membrane (hAM). hAM is a transparent protective membrane that surrounds the fetus. Preclinical studies showed pro-apoptotic and antiproliferative properties of hAM treatment on cancer cells. Herein, we present the latest findings of the application of the hAM in combating HCC tumorigenesis and the underlying molecular pathogenies and the role of transforming growth factor-beta (TGFβ), P53, WNT/beta-catenin, and PI3K/AKT pathways. The emerging clinical applications of hAM in cancer therapy provide evidence for its diverse and unique features and suitability for the management of a wide range of pathological conditions.
Collapse
Affiliation(s)
- Ahmed M Abou-Shanab
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, Egypt
| | - Ola A Gaser
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, Egypt
| | - Radwa Ayman Salah
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, Egypt.
| |
Collapse
|
35
|
Harland N, Knoll J, Amend B, Abruzzese T, Abele H, Jakubowski P, Stenzl A, Aicher WK. Xenogenic Application of Human Placenta-Derived Mesenchymal Stromal Cells in a Porcine Large Animal Model. Cell Transplant 2024; 33:9636897241226737. [PMID: 38323325 PMCID: PMC10851762 DOI: 10.1177/09636897241226737] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/30/2023] [Accepted: 01/02/2024] [Indexed: 02/08/2024] Open
Abstract
In animal models, cell therapies for different diseases or injuries have been very successful. Preclinical studies with cells aiming at a stroke, heart attack, and other emergency situations were promising but sometimes failed translation in clinical situations. We, therefore, investigated if human placenta-derived mesenchymal stromal cells can be injected in pigs without provoking rejection to serve as a xenogenic transplantation model to bridge preclinical animal studies to more promising future preclinical studies. Male human placenta-derived mesenchymal stromal cells were isolated, expanded, and characterized by flow cytometry, in vitro differentiation, and quantitative reverse-transcription polymerase chain reaction to prove their nature. Such cells were injected into the sphincter muscle of the urethrae of female pigs under visual control by cystoscopy employing a Williams needle. The animals were observed over 7 days of follow-up. Reactions of the host to the xenogeneic cells were explored by monitoring body temperature, and inflammatory markers including IL-1ß, CRP, and haptoglobin in blood. After sacrifice on day 7, infiltration of inflammatory cells in the tissue targeted was investigated by histology and immunofluorescence. DNA of injected human cells was detected by PCR. Upon injection in vascularized porcine tissue, human placenta-derived mesenchymal stromal cells were tolerated, and systemic inflammatory parameters were not elevated. DNA of injected cells was detected in situ 7 days after injection, and moderate local infiltration of inflammatory cells was observed. The therapeutic potential of human placenta-derived mesenchymal stromal cells can be explored in porcine large animal models of injury or disease. This seems a promising strategy to explore technologies for cell injections in infarcted hearts or small organs and tissues in therapeutically relevant amounts requiring large animal models to yield meaningful outcomes.
Collapse
Affiliation(s)
- Niklas Harland
- Department of Urology, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Jasmin Knoll
- Center for Medical Research, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Bastian Amend
- Department of Urology, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Tanja Abruzzese
- Center for Medical Research, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Harald Abele
- Department of Gynecology and Obstetrics, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Peter Jakubowski
- Department of Gynecology and Obstetrics, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Arnulf Stenzl
- Center for Medical Research, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Wilhelm K. Aicher
- Department of Urology, University Hospital, Eberhard Karls University, Tuebingen, Germany
| |
Collapse
|
36
|
Paul M, Ain R. Evaluation of Molecular Interactions and Cellular Dynamics at the Maternal-Fetal Interface During Placental Morphogenesis. Methods Mol Biol 2024; 2728:45-76. [PMID: 38019391 DOI: 10.1007/978-1-0716-3495-0_5] [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] [Indexed: 11/30/2023]
Abstract
Molecular events at the maternal-fetal interface establish successful pregnancies. Identifying and characterizing the heterogeneous cell population and their cross-talk at the cellular and molecular levels are essential to expand our knowledge on the progression and maintenance of pregnancy. In this chapter, we briefly discuss the organization of maternal-fetal interface in mice/rats and humans. We illustrate methods for studying the cell composition using flow cytometry, immunocytochemical and biochemical studies, intercellular interaction using co-culture system and spheroid assay, and function of trophoblast cells using ELISA, RNA sequencing, mass spectrometry (MS) to analyze the proteome, invasion assay, and scratch wound assay.
Collapse
Affiliation(s)
- Madhurima Paul
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Rupasri Ain
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India.
| |
Collapse
|
37
|
de Laorden EH, Simón D, Milla S, Portela-Lomba M, Mellén M, Sierra J, de la Villa P, Moreno-Flores MT, Iglesias M. Human placenta-derived mesenchymal stem cells stimulate neuronal regeneration by promoting axon growth and restoring neuronal activity. Front Cell Dev Biol 2023; 11:1328261. [PMID: 38188022 PMCID: PMC10766706 DOI: 10.3389/fcell.2023.1328261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/11/2023] [Indexed: 01/09/2024] Open
Abstract
In the last decades, mesenchymal stem cells (MSCs) have become the cornerstone of cellular therapy due to their unique characteristics. Specifically human placenta-derived mesenchymal stem cells (hPMSCs) are highlighted for their unique features, including ease to isolate, non-invasive techniques for large scale cell production, significant immunomodulatory capacity, and a high ability to migrate to injuries. Researchers are exploring innovative techniques to overcome the low regenerative capacity of Central Nervous System (CNS) neurons, with one promising avenue being the development of tailored mesenchymal stem cell therapies capable of promoting neural repair and recovery. In this context, we have evaluated hPMSCs as candidates for CNS lesion regeneration using a skillful co-culture model system. Indeed, we have demonstrated the hPMSCs ability to stimulate damaged rat-retina neurons regeneration by promoting axon growth and restoring neuronal activity both under normoxia and hypoxia conditions. With our model we have obtained neuronal regeneration values of 10%-14% and axonal length per neuron rates of 19-26, μm/neuron. To assess whether the regenerative capabilities of hPMSCs are contact-dependent effects or it is mediated through paracrine mechanisms, we carried out transwell co-culture and conditioned medium experiments confirming the role of secreted factors in axonal regeneration. It was found that hPMSCs produce brain derived, neurotrophic factor (BDNF), nerve-growth factor (NGF) and Neurotrophin-3 (NT-3), involved in the process of neuronal regeneration and restoration of the physiological activity of neurons. In effect, we confirmed the success of our treatment using the patch clamp technique to study ionic currents in individual isolated living cells demonstrating that in our model the regenerated neurons are electrophysiologically active, firing action potentials. The outcomes of our neuronal regeneration studies, combined with the axon-regenerating capabilities exhibited by mesenchymal stem cells derived from the placenta, present a hopeful outlook for the potential therapeutic application of hPMSCs in the treatment of neurological disorders.
Collapse
Affiliation(s)
- Elvira H. de Laorden
- Facultad de C.C. Experimentales, Universidad Francisco de Vitoria, Madrid, Spain
| | - Diana Simón
- Facultad de C.C. Experimentales, Universidad Francisco de Vitoria, Madrid, Spain
| | - Santiago Milla
- Departamento de Biología de Sistemas, Unidad de Fisiología, Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá, Alcalá de Henares, Spain
| | - María Portela-Lomba
- Facultad de C.C. Experimentales, Universidad Francisco de Vitoria, Madrid, Spain
| | - Marian Mellén
- Facultad de C.C. Experimentales, Universidad Francisco de Vitoria, Madrid, Spain
| | - Javier Sierra
- Facultad de C.C. Experimentales, Universidad Francisco de Vitoria, Madrid, Spain
| | - Pedro de la Villa
- Departamento de Biología de Sistemas, Unidad de Fisiología, Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá, Alcalá de Henares, Spain
| | - María Teresa Moreno-Flores
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Maite Iglesias
- Facultad de C.C. Experimentales, Universidad Francisco de Vitoria, Madrid, Spain
| |
Collapse
|
38
|
Fioretti D, Ledda M, Iurescia S, Carletti R, Di Gioia C, Lolli MG, Marchese R, Lisi A, Rinaldi M. Severely Damaged Freeze-Injured Skeletal Muscle Reveals Functional Impairment, Inadequate Repair, and Opportunity for Human Stem Cell Application. Biomedicines 2023; 12:30. [PMID: 38275391 PMCID: PMC10813063 DOI: 10.3390/biomedicines12010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND The regeneration of severe traumatic muscle injuries is an unsolved medical need that is relevant for civilian and military medicine. In this work, we produced a critically sized nonhealing muscle defect in a mouse model to investigate muscle degeneration/healing phases. MATERIALS AND METHODS We caused a freeze injury (FI) in the biceps femoris of C57BL/6N mice. From day 1 to day 25 post-injury, we conducted histological/morphometric examinations, an analysis of the expression of genes involved in inflammation/regeneration, and an in vivo functional evaluation. RESULTS We found that FI activates cytosolic DNA sensing and inflammatory responses. Persistent macrophage infiltration, the prolonged expression of eMHC, the presence of centrally nucleated myofibers, and the presence of PAX7+ satellite cells at late time points and with chronic physical impairment indicated inadequate repair. By looking at stem-cell-based therapeutic protocols of muscle repair, we investigated the crosstalk between M1-biased macrophages and human amniotic mesenchymal stem cells (hAMSCs) in vitro. We demonstrated their reciprocal paracrine effects where hAMSCs induced a shift of M1 macrophages into an anti-inflammatory phenotype, and M1 macrophages promoted an increase in the expression of hAMSC immunomodulatory factors. CONCLUSIONS Our findings support the rationale for the future use of our injury model to exploit the full potential of in vivo hAMSC transplantation following severe traumatic injuries.
Collapse
Affiliation(s)
- Daniela Fioretti
- Department Biomedical Sciences, Institute of Translational Pharmacology, National Research Council, Area di Ricerca Roma2 Tor Vergata, 00133 Rome, Italy; (M.L.); (S.I.); (M.G.L.); (A.L.)
| | - Mario Ledda
- Department Biomedical Sciences, Institute of Translational Pharmacology, National Research Council, Area di Ricerca Roma2 Tor Vergata, 00133 Rome, Italy; (M.L.); (S.I.); (M.G.L.); (A.L.)
| | - Sandra Iurescia
- Department Biomedical Sciences, Institute of Translational Pharmacology, National Research Council, Area di Ricerca Roma2 Tor Vergata, 00133 Rome, Italy; (M.L.); (S.I.); (M.G.L.); (A.L.)
| | - Raffaella Carletti
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy;
| | - Cira Di Gioia
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, 00161 Rome, Italy;
| | - Maria Grazia Lolli
- Department Biomedical Sciences, Institute of Translational Pharmacology, National Research Council, Area di Ricerca Roma2 Tor Vergata, 00133 Rome, Italy; (M.L.); (S.I.); (M.G.L.); (A.L.)
| | - Rodolfo Marchese
- Department of Clinical Pathology, FBF S. Peter Hospital, 00189 Rome, Italy;
| | - Antonella Lisi
- Department Biomedical Sciences, Institute of Translational Pharmacology, National Research Council, Area di Ricerca Roma2 Tor Vergata, 00133 Rome, Italy; (M.L.); (S.I.); (M.G.L.); (A.L.)
| | - Monica Rinaldi
- Department Biomedical Sciences, Institute of Translational Pharmacology, National Research Council, Area di Ricerca Roma2 Tor Vergata, 00133 Rome, Italy; (M.L.); (S.I.); (M.G.L.); (A.L.)
| |
Collapse
|
39
|
Jantalika T, Manochantr S, Kheolamai P, Tantikanlayaporn D, Thongsepee N, Warnnissorn N, Saijuntha W, Pinlaor S, Tantrawatpan C. The Human Placental Amniotic Membrane Mesenchymal-Stromal-Cell-Derived Conditioned Medium Inhibits Growth and Promotes Apoptosis of Human Cholangiocarcinoma Cells In Vitro and In Vivo by Suppressing IL-6/JAK2/STAT3 Signaling. Cells 2023; 12:2788. [PMID: 38132108 PMCID: PMC10742162 DOI: 10.3390/cells12242788] [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: 10/12/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) have recently been shown to play an important role in the growth and progression of many solid tumors, including cholangiocarcinoma (CCA). The human placental amniotic membrane (hPAM) is one of the most favorable sources of MSCs due to its availability and non-invasive harvesting procedure. However, the role of human placental amniotic membrane mesenchymal stromal cells (hPAMSCs) in the growth and progression of human CCA has not yet been determined. This study investigates the effects of conditioned medium derived from hPAMSCs (PA-CM) on the properties of three human CCA cell lines and explores possible mechanisms of action. Varying concentrations of PA-CM were used to treat CCA cells to determine their effects on the proliferation and apoptosis of CCA cells. The results showed that PA-CM inhibited the proliferation and colony-forming capacity of KKU100, KKU213A, and KKU213B cells. PA-CM also promoted the apoptosis of these CCA cells by causing the loss of mitochondrial membrane potential. Western Blotting confirmed that PA-CM induced CCA cell apoptosis by increasing the levels of the Bax/Bcl-2 ratio, cleaved caspase 3, and cleaved PARP, possibly by inhibiting the IL-6/JAK2/STAT3 signaling pathway. Moreover, our in vivo study also confirmed the suppressive effect of hPAMSCs on CCA cells by showing that PA-CM reduced tumor volume in nude mice transplanted with human CCA cells. Taken together, our results demonstrate that PA-CM has potent tumor-suppressive effects on human CCA cells and could potentially be used in combination with chemotherapy to develop a more effective treatment for CCA patients.
Collapse
Affiliation(s)
- Tanachapa Jantalika
- Division of Cell Biology, Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (T.J.); (S.M.); (P.K.); (D.T.)
- Center of Excellence in Stem Cell Research and Innovations, Thammasat University, Pathum Thani 12120, Thailand
| | - Sirikul Manochantr
- Division of Cell Biology, Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (T.J.); (S.M.); (P.K.); (D.T.)
- Center of Excellence in Stem Cell Research and Innovations, Thammasat University, Pathum Thani 12120, Thailand
| | - Pakpoom Kheolamai
- Division of Cell Biology, Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (T.J.); (S.M.); (P.K.); (D.T.)
- Center of Excellence in Stem Cell Research and Innovations, Thammasat University, Pathum Thani 12120, Thailand
| | - Duangrat Tantikanlayaporn
- Division of Cell Biology, Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (T.J.); (S.M.); (P.K.); (D.T.)
- Center of Excellence in Stem Cell Research and Innovations, Thammasat University, Pathum Thani 12120, Thailand
| | - Nattaya Thongsepee
- Division of Physiology, Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathum Thani 12120, Thailand;
- Research Unit in Nutraceuticals and Food Safety, Thammasat University, Pathum Thani 12120, Thailand
| | - Naree Warnnissorn
- Department of Pathology, Faculty of Medicine, Thammasat University, Pathum Thani 12120, Thailand;
| | - Weerachai Saijuntha
- Faculty of Medicine, Mahasarakham University, Maha Sarakham 44000, Thailand;
| | - Somchai Pinlaor
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Chairat Tantrawatpan
- Division of Cell Biology, Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (T.J.); (S.M.); (P.K.); (D.T.)
- Center of Excellence in Stem Cell Research and Innovations, Thammasat University, Pathum Thani 12120, Thailand
| |
Collapse
|
40
|
Bačenková D, Trebuňová M, Demeterová J, Živčák J. Human Chondrocytes, Metabolism of Articular Cartilage, and Strategies for Application to Tissue Engineering. Int J Mol Sci 2023; 24:17096. [PMID: 38069417 PMCID: PMC10707713 DOI: 10.3390/ijms242317096] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Hyaline cartilage, which is characterized by the absence of vascularization and innervation, has minimal self-repair potential in case of damage and defect formation in the chondral layer. Chondrocytes are specialized cells that ensure the synthesis of extracellular matrix components, namely type II collagen and aggregen. On their surface, they express integrins CD44, α1β1, α3β1, α5β1, α10β1, αVβ1, αVβ3, and αVβ5, which are also collagen-binding components of the extracellular matrix. This article aims to contribute to solving the problem of the possible repair of chondral defects through unique methods of tissue engineering, as well as the process of pathological events in articular cartilage. In vitro cell culture models used for hyaline cartilage repair could bring about advanced possibilities. Currently, there are several variants of the combination of natural and synthetic polymers and chondrocytes. In a three-dimensional environment, chondrocytes retain their production capacity. In the case of mesenchymal stromal cells, their favorable ability is to differentiate into a chondrogenic lineage in a three-dimensional culture.
Collapse
Affiliation(s)
- Darina Bačenková
- Department of Biomedical Engineering and Measurement, Faculty of Mechanical Engineering, Technical University of Košice, Letná 9, 042 00 Košice, Slovakia; (M.T.); (J.D.); (J.Ž.)
| | | | | | | |
Collapse
|
41
|
Lee C, Liao Z, Li Y, Lai Q, Guo Y, Huang J, Li S, Wang Y, Shi R. Placental MRI segmentation based on multi-receptive field and mixed attention separation mechanism. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 242:107699. [PMID: 37769416 DOI: 10.1016/j.cmpb.2023.107699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 06/21/2023] [Accepted: 06/25/2023] [Indexed: 09/30/2023]
Abstract
OBJECTIVE To reduce the occurrence of massive bleeding during placental abruption in patients with placenta accrete, we established a medical imaging based on multi-receptive field and mixed attention separation mechanism (MRF-MAS) model to improve the accuracy of MRI placenta segmentation and provide a basis for subsequent placenta accreta. METHODS We propose a placenta MRI segmentation technology using the MRF-MAS framework to develop a medical image diagnostic technique. The model first uses the multi-receptive field feature structure to obtain multi-level information, and improves the expression of features at differing scales. Note that the hybrid attention mechanism combines channel attention and spatial attention, separates the input feature sets and computes the attention separately, and finally reorganizes the feature maps. To show that the model can improve the accuracy of segmenting the placenta, we adopt mean Intersection over Union (IoU), Dice similarity coefficient (Dice) and area under the receiver operating characteristic curve (AUC) with U-Net, Mask RCNN, Deeplab v3 for comparison. RESULTS The four models achieved different outcomes based on our placenta dataset, with our model IoU and Dice up to 0.8169 and 0.8992, which are 5.51% and 3.03% higher than the average of the three comparison models. CONCLUSION The model proposed by us is helpful to assist the imaging diagnosis and at the same time provides a quantitative reference for the precise treatment of placenta accreta, assists the Equationtion of the clinical operation plan of the physician, and promotes the precision medicine of placenta accreta.
Collapse
Affiliation(s)
- Cong Lee
- School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Zhifang Liao
- School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Yuanzhe Li
- Department of CT/MRI, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Qingquan Lai
- Department of CT/MRI, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Yingying Guo
- Department of CT/MRI, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Jing Huang
- Department of CT/MRI, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Shuting Li
- Department of CT/MRI, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Yi Wang
- Department of CT/MRI, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Ruizheng Shi
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
42
|
Muntiu A, Papait A, Vincenzoni F, Vitali A, Lattanzi W, Romele P, Cargnoni A, Silini A, Parolini O, Desiderio C. Disclosing the molecular profile of the human amniotic mesenchymal stromal cell secretome by filter-aided sample preparation proteomic characterization. Stem Cell Res Ther 2023; 14:339. [PMID: 38012707 PMCID: PMC10683150 DOI: 10.1186/s13287-023-03557-4] [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: 01/20/2023] [Accepted: 10/30/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND The secretome of mesenchymal stromal cells isolated from the amniotic membrane (hAMSCs) has been extensively studied for its in vitro immunomodulatory activity as well as for the treatment of several preclinical models of immune-related disorders. The bioactive molecules within the hAMSCs secretome are capable of modulating the immune response and thus contribute to stimulating regenerative processes. At present, only a few studies have attempted to define the composition of the secretome, and several approaches, including multi-omics, are underway in an attempt to precisely define its composition and possibly identify key factors responsible for the therapeutic effect. METHODS In this study, we characterized the protein composition of the hAMSCs secretome by a filter-aided sample preparation (FASP) digestion and liquid chromatography-high resolution mass spectrometry (LC-MS) approach. Data were processed for gene ontology classification and functional protein interaction analysis by bioinformatics tools. RESULTS Proteomic analysis of the hAMSCs secretome resulted in the identification of 1521 total proteins, including 662 unique elements. A number of 157 elements, corresponding to 23.7%, were found as repeatedly characterizing the hAMSCs secretome, and those that resulted as significantly over-represented were involved in immunomodulation, hemostasis, development and remodeling of the extracellular matrix molecular pathways. CONCLUSIONS Overall, our characterization enriches the landscape of hAMSCs with new information that could enable a better understanding of the mechanisms of action underlying the therapeutic efficacy of the hAMSCs secretome while also providing a basis for its therapeutic translation.
Collapse
Affiliation(s)
- Alexandra Muntiu
- Istituto di Scienze e Tecnologie Chimiche (SCITEC) ''Giulio Natta'', Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Andrea Papait
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario ''Agostino Gemelli'' Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Rome, Italy
| | - Federica Vincenzoni
- Fondazione Policlinico Universitario ''Agostino Gemelli'' Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Rome, Italy
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alberto Vitali
- Istituto di Scienze e Tecnologie Chimiche (SCITEC) ''Giulio Natta'', Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Wanda Lattanzi
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario ''Agostino Gemelli'' Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Rome, Italy
| | - Pietro Romele
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Anna Cargnoni
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Antonietta Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy.
- Fondazione Policlinico Universitario ''Agostino Gemelli'' Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Rome, Italy.
| | - Claudia Desiderio
- Istituto di Scienze e Tecnologie Chimiche (SCITEC) ''Giulio Natta'', Consiglio Nazionale delle Ricerche, Rome, Italy.
| |
Collapse
|
43
|
Jahangiri B, Khalaj-Kondori M, Asadollahi E, Kian Saei A, Sadeghizadeh M. Dual impacts of mesenchymal stem cell-derived exosomes on cancer cells: unravelling complex interactions. J Cell Commun Signal 2023:10.1007/s12079-023-00794-3. [PMID: 37973719 DOI: 10.1007/s12079-023-00794-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent, self-renewing stromal cells found in a variety of adult tissues. MSCs possess a remarkable ability to migrate towards tumor sites, known as homing. This homing process is mediated by various factors, including chemokines, growth factors, and extracellular matrix components present in the tumor microenvironment. MSCs release extracellular vesicles known as exosomes (MSC-Exos), which have been suggested to serve a key role in mediating a wide variety of MSC activities. Through cell-cell communication, MSC-Exos have been shown to alter recipient cell phenotype or function and play as a novel cell-free alternative for MSC-based cell therapy. However, MSC recruitment to tumors allows for their interaction with cancer cells and subsequent regulation of tumor behavior. MSC-Exos act as tumor niche modulators via transferring exosomal contents, such as specific proteins or genetic materials, to the nearby cancer cells, leading to either promotion or suppression of tumorigenesis, angiogenesis, and metastasis, depending on the specific microenvironmental cues and recipient cell characteristics. Consequently, there is still a debate about the precise relationship between tumor cells and MSC-Exos, and it is unclear how MSC-Exos impacts tumor cells. Although the dysregulation of miRNAs is caused by the progression of cancer, they also play a direct role in either promoting or inhibiting tumor growth as they act as either oncogenes or tumor suppressors. The utilization of MSC-Exos may prove to be an effective method for restoring miRNA as a means of treating cancer. This review aimed to present the existing understanding of the impact that MSC-Exos could have on cancer. To begin with, we presented a brief explanation of exosomes, MSCs, and MSC-Exos. Following this, we delved into the impact of MSC-Exos on cancer growth, EMT, metastasis, angiogenesis, resistance to chemotherapy and radiotherapy, and modulation of the immune system. Opposing effects of mesenchymal stem cells-derived exosomes on cancer cells.
Collapse
Affiliation(s)
- Babak Jahangiri
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | - Elahe Asadollahi
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ali Kian Saei
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Majid Sadeghizadeh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
44
|
Amend B, Buttgereit L, Abruzzese T, Harland N, Abele H, Jakubowski P, Stenzl A, Gorodetsky R, Aicher WK. Regulation of Immune Checkpoint Antigen CD276 (B7-H3) on Human Placenta-Derived Mesenchymal Stromal Cells in GMP-Compliant Cell Culture Media. Int J Mol Sci 2023; 24:16422. [PMID: 38003612 PMCID: PMC10671289 DOI: 10.3390/ijms242216422] [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: 10/09/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Therapies utilizing autologous mesenchymal cell delivery are being investigated as anti-inflammatory and regenerative treatments for a broad spectrum of age-related diseases, as well as various chronic and acute pathological conditions. Easily available allogeneic full-term human placenta mesenchymal stromal cells (pMSCs) were used as a potential pro-regenerative, cell-based therapy in degenerative diseases, which could be applied also to elderly individuals. To explore the potential of allogeneic pMSCs transplantation for pro-regenerative applications, such cells were isolated from five different term-placentas, obtained from the dissected maternal, endometrial (mpMSCs), and fetal chorion tissues (fpMSCs), respectively. The proliferation rate of the cells in the culture, as well as their shape, in vitro differentiation potential, and the expression of mesenchymal lineage and stem cell markers, were investigated. Moreover, we studied the expression of immune checkpoint antigen CD276 as a possible modulation of the rejection of transplanted non-HLA-matched homologous or even xeno-transplanted pMSCs. The expression of the cell surface markers was also explored in parallel in the cryosections of the relevant intact placenta tissue samples. The expansion of pMSCs in a clinical-grade medium complemented with 5% human platelet lysate and 5% human serum induced a significant expression of CD276 when compared to mpMSCs expanded in a commercial medium. We suggest that the expansion of mpMSCs, especially in a medium containing platelet lysate, elevated the expression of the immune-regulatory cell surface marker CD276. This may contribute to the immune tolerance towards allogeneic pMSC transplantations in clinical situations and even in xenogenic animal models of human diseases. The endurance of the injected comparably young human-term pMSCs may promote prolonged effects in clinical applications employing non-HLA-matched allogeneic cell therapy for various degenerative disorders, especially in aged adults.
Collapse
Affiliation(s)
- Bastian Amend
- Department of Urology, University Hospital, Eberhard Karls University, 72076 Tuebingen, Germany
| | - Lea Buttgereit
- Centre for Medical Research, Department of Urology, Eberhard Karls University, 72076 Tuebingen, Germany
| | - Tanja Abruzzese
- Centre for Medical Research, Department of Urology, Eberhard Karls University, 72076 Tuebingen, Germany
| | - Niklas Harland
- Department of Urology, University Hospital, Eberhard Karls University, 72076 Tuebingen, Germany
| | - Harald Abele
- Department of Gynaecology and Obstetrics, University Hospital, Eberhard Karls University, 72076 Tuebingen, Germany
| | - Peter Jakubowski
- Department of Gynaecology and Obstetrics, University Hospital, Eberhard Karls University, 72076 Tuebingen, Germany
| | - Arnulf Stenzl
- Department of Urology, University Hospital, Eberhard Karls University, 72076 Tuebingen, Germany
| | - Raphael Gorodetsky
- Biotechnology and Radiobiology Laboratory, Sharett Institute of Oncology, Hadassah-Hebrew University Medical Centre, Jerusalem 91120, Israel
| | - Wilhelm K. Aicher
- Centre for Medical Research, Department of Urology, Eberhard Karls University, 72076 Tuebingen, Germany
| |
Collapse
|
45
|
Lee CM, Go YY, Song JJ. Inhibition of lipopolysaccharide-induced inflammation by trophoblast-conditioned medium and trophoblast-derived extracellular vesicles in human middle ear epithelial cells. Sci Rep 2023; 13:19822. [PMID: 37963902 PMCID: PMC10645728 DOI: 10.1038/s41598-023-46731-7] [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: 05/19/2023] [Accepted: 11/04/2023] [Indexed: 11/16/2023] Open
Abstract
Otitis media is a common disease but can cause severe inner ear inflammation and hearing loss if it persists for more than two weeks. This study elucidates the inflammation-inhibiting efficacy of conditioned medium (CM) and extracellular vesicles (EVs) derived from human trophoblast (TB) cells in lipopolysaccharide (LPS)-induced human middle ear epithelial cells (HMEECs). TB-conditioned medium (TB-CM) reduced the inflammatory response and regulated mucin and epithelial sodium channel genes in LPS-induced HMEECs. The underlying mechanism of cell migration during inflammatory healing in LPS-induced HMEECs treated with TB-CM was determined by RNA-sequencing analysis. Specifically, the NF-κB pathway related to the copper metabolism MURR1 domain protein was studied and verified through siRNA. This elucidation of the anti-inflammatory effect of TB-CM and TB-derived EVs demonstrates their clinical potential to treat chronic inflammation.
Collapse
Affiliation(s)
- Chan Mi Lee
- Division of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yoon Young Go
- Division of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
- Center for Health Care Convergence at Korea University Guro Hospital, Seoul, Republic of Korea
| | - Jae-Jun Song
- Division of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea.
- Center for Health Care Convergence at Korea University Guro Hospital, Seoul, Republic of Korea.
| |
Collapse
|
46
|
Ingraldi AL, Audet RG, Tabor AJ. The Preparation and Clinical Efficacy of Amnion-Derived Membranes: A Review. J Funct Biomater 2023; 14:531. [PMID: 37888195 PMCID: PMC10607219 DOI: 10.3390/jfb14100531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023] Open
Abstract
Biological tissues from various anatomical sources have been utilized for tissue transplantation and have developed into an important source of extracellular scaffolding material for regenerative medicine applications. Tissue scaffolds ideally integrate with host tissue and provide a homeostatic environment for cellular infiltration, growth, differentiation, and tissue resolution. The human amniotic membrane is considered an important source of scaffolding material due to its 3D structural architecture and function and as a source of growth factors and cytokines. This tissue source has been widely studied and used in various areas of tissue repair including intraoral reconstruction, corneal repair, tendon repair, microvascular reconstruction, nerve procedures, burns, and chronic wound treatment. The production of amniotic membrane allografts has not been standardized, resulting in a wide array of amniotic membrane products, including single, dual, and tri-layered products, such as amnion, chorion, amnion-chorion, amnion-amnion, and amnion-chorion-amnion allografts. Since these allografts are not processed using the same methods, they do not necessarily produce the same clinical responses. The aim of this review is to highlight the properties of different human allograft membranes, present the different processing and preservation methods, and discuss their use in tissue engineering and regenerative applications.
Collapse
Affiliation(s)
- Alison L. Ingraldi
- Carmell Corporation, Pittsburg, PA 15203, USA;
- Department of Research and Development, Axolotl Biologix, Flagstaff, AZ 86001, USA
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Robert G. Audet
- Carmell Corporation, Pittsburg, PA 15203, USA;
- Department of Research and Development, Axolotl Biologix, Flagstaff, AZ 86001, USA
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Aaron J. Tabor
- Carmell Corporation, Pittsburg, PA 15203, USA;
- Department of Research and Development, Axolotl Biologix, Flagstaff, AZ 86001, USA
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ 86011, USA
- Department of Clinical Operations, Axolotl Biologix, Flagstaff, AZ 86001, USA
| |
Collapse
|
47
|
Hu Z, Luo Y, Ni R, Hu Y, Yang F, Du T, Zhu Y. Biological importance of human amniotic membrane in tissue engineering and regenerative medicine. Mater Today Bio 2023; 22:100790. [PMID: 37711653 PMCID: PMC10498009 DOI: 10.1016/j.mtbio.2023.100790] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 08/21/2023] [Accepted: 08/31/2023] [Indexed: 09/16/2023] Open
Abstract
The human amniotic membrane (hAM) is the innermost layer of the placenta. Its distinctive structure and the biological and physical characteristics make it a highly biocompatible material in a variety of regenerative medicine applications. It also acts as a supply of bioactive factors and cells, which indicate the advantages over other tissues. In this review, we firstly discussed the biological properties of hAM-derived cells in vivo or in vitro, along with their stemness of markers, pointing out a promising source of stem cells for regenerative medicine. Then, we systematically summarized current knowledge on the collection, preparation, preservation, and decellularization of hAM, as well as their characteristics helping to improve the understanding of applications in tissue engineering. Finally, we highlighted the recent advances in which hAM has undergone additional modifications to achieve an adequate perspective of regenerative medicine applications. More investigations are required in utilizing appropriate modifications to enhance the therapeutic effectiveness of hAM in the future.
Collapse
Affiliation(s)
- Zeming Hu
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yang Luo
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Renhao Ni
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yiwei Hu
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Fang Yang
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Tianyu Du
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yabin Zhu
- Health Science Center, Ningbo University, Ningbo, 315211, China
| |
Collapse
|
48
|
Mizraji G, Davidzohn A, Gursoy M, Gursoy U, Shapira L, Wilensky A. Membrane barriers for guided bone regeneration: An overview of available biomaterials. Periodontol 2000 2023; 93:56-76. [PMID: 37855164 DOI: 10.1111/prd.12502] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/21/2023] [Accepted: 05/29/2023] [Indexed: 10/20/2023]
Abstract
Dental implants revolutionized the treatment options for restoring form, function, and esthetics when one or more teeth are missing. At sites of insufficient bone, guided bone regeneration (GBR) is performed either prior to or in conjunction with implant placement to achieve a three-dimensional prosthetic-driven implant position. To date, GBR is well documented, widely used, and constitutes a predictable and successful approach for lateral and vertical bone augmentation of atrophic ridges. Evidence suggests that the use of barrier membranes maintains the major biological principles of GBR. Since the material used to construct barrier membranes ultimately dictates its characteristics and its ability to maintain the biological principles of GBR, several materials have been used over time. This review, summarizes the evolution of barrier membranes, focusing on the characteristics, advantages, and disadvantages of available occlusive barrier membranes and presents results of updated meta-analyses focusing on the effects of these membranes on the overall outcome.
Collapse
Affiliation(s)
- Gabriel Mizraji
- Department of Periodontology, Faculty of Dental Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Mervi Gursoy
- Department of Periodontology, Institute of Dentistry, University of Turku, Turku, Finland
- Oral Health Care, Welfare Division, City of Turku, Turku, Finland
| | - Ulvi Gursoy
- Department of Periodontology, Institute of Dentistry, University of Turku, Turku, Finland
| | - Lior Shapira
- Department of Periodontology, Faculty of Dental Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Asaf Wilensky
- Department of Periodontology, Faculty of Dental Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
49
|
Russo E, Alberti G, Corrao S, Borlongan CV, Miceli V, Conaldi PG, Di Gaudio F, La Rocca G. The Truth Is Out There: Biological Features and Clinical Indications of Extracellular Vesicles from Human Perinatal Stem Cells. Cells 2023; 12:2347. [PMID: 37830562 PMCID: PMC10571796 DOI: 10.3390/cells12192347] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 10/14/2023] Open
Abstract
The potential of perinatal tissues to provide cellular populations to be used in different applications of regenerative medicine is well established. Recently, the efforts of researchers are being addressed regarding the evaluation of cell products (secreted molecules or extracellular vesicles, EVs) to be used as an alternative to cellular infusion. The data regarding the effective recapitulation of most perinatal cells' properties by their secreted complement point in this direction. EVs secreted from perinatal cells exhibit key therapeutic effects such as tissue repair and regeneration, the suppression of inflammatory responses, immune system modulation, and a variety of other functions. Although the properties of EVs from perinatal derivatives and their significant potential for therapeutic success are amply recognized, several challenges still remain that need to be addressed. In the present review, we provide an up-to-date analysis of the most recent results in the field, which can be addressed in future research in order to overcome the challenges that are still present in the characterization and utilization of the secreted complement of perinatal cells and, in particular, mesenchymal stromal cells.
Collapse
Affiliation(s)
- Eleonora Russo
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (E.R.); (G.A.)
| | - Giusi Alberti
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (E.R.); (G.A.)
| | - Simona Corrao
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (S.C.); (V.M.); (P.G.C.)
| | - Cesar V. Borlongan
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL 33620, USA;
| | - Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (S.C.); (V.M.); (P.G.C.)
| | - Pier Giulio Conaldi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (S.C.); (V.M.); (P.G.C.)
| | - Francesca Di Gaudio
- Department of Health Promotion, Maternal-Infantile Care, Excellence Internal and Specialist Medicine “G. D’Alessandro” (PROMISE), University of Palermo, 90127 Palermo, Italy;
| | - Giampiero La Rocca
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (E.R.); (G.A.)
| |
Collapse
|
50
|
Vaiasicca S, Melone G, James DW, Quintela M, Preziuso A, Finnell RH, Conlan RS, Francis LW, Corradetti B. Transcriptomic analysis of stem cells from chorionic villi uncovers the impact of chromosomes 2, 6 and 22 in the clinical manifestations of Down syndrome. Stem Cell Res Ther 2023; 14:265. [PMID: 37740230 PMCID: PMC10517537 DOI: 10.1186/s13287-023-03503-4] [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: 04/10/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND Down syndrome (DS) clinical multisystem condition is generally considered the result of a genetic imbalance generated by the extra copy of chromosome 21. Recent discoveries, however, demonstrate that the molecular mechanisms activated in DS compared to euploid individuals are more complex than previously thought. Here, we utilize mesenchymal stem cells from chorionic villi (CV) to uncover the role of comprehensive functional genomics-based understanding of DS complexity. METHODS Next-generation sequencing coupled with bioinformatic analysis was performed on CV obtained from women carrying fetuses with DS (DS-CV) to reveal specific genome-wide transcriptional changes compared to their euploid counterparts. Functional assays were carried out to confirm the biological processes identified as enriched in DS-CV compared to CV (i.e., cell cycle, proliferation features, immunosuppression and ROS production). RESULTS Genes located on chromosomes other than the canonical 21 (Ch. 2, 6 and 22) are responsible for the impairment of life-essential pathways, including cell cycle regulation, innate immune response and reaction to external stimuli were found to be differentially expressed in DS-CV. Experimental validation confirmed the key role of the biological pathways regulated by those genes in the etiology of such a multisystem condition. CONCLUSIONS NGS dataset generated in this study highlights the compromised functionality in the proliferative rate and in the innate response of DS-associated clinical conditions and identifies DS-CV as suitable tools for the development of specifically tailored, personalized intervention modalities.
Collapse
Affiliation(s)
- Salvatore Vaiasicca
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
- Scientific Direction, IRCCS INRCA, Ancona, Italy
| | - Gianmarco Melone
- Centre for NanoHealth, Swansea University Medical School, Singleton Park, Swansea, Wales, UK
| | - David W James
- Centre for NanoHealth, Swansea University Medical School, Singleton Park, Swansea, Wales, UK
| | - Marcos Quintela
- Centre for NanoHealth, Swansea University Medical School, Singleton Park, Swansea, Wales, UK
| | - Alessandra Preziuso
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Richard H Finnell
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Robert Steven Conlan
- Centre for NanoHealth, Swansea University Medical School, Singleton Park, Swansea, Wales, UK
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Lewis W Francis
- Centre for NanoHealth, Swansea University Medical School, Singleton Park, Swansea, Wales, UK
| | - Bruna Corradetti
- Centre for NanoHealth, Swansea University Medical School, Singleton Park, Swansea, Wales, UK.
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA.
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|