1
|
Ait Hamdan Y, El-Mansoury B, Elouali S, Rachmoune K, Belbachir A, Oudadesse H, Rhazi M. A review of chitosan polysaccharides: Neuropharmacological implications and tissue regeneration. Int J Biol Macromol 2024; 279:135356. [PMID: 39244136 DOI: 10.1016/j.ijbiomac.2024.135356] [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: 06/12/2024] [Revised: 08/20/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
One of the current challenges in targeting neurological disorders is that many therapeutic molecules cannot cross the blood-brain barrier (BBB), which limits the use of natural molecules in nervous tissue regeneration. Thus, the development of new drugs to effectively treat neurological disorders would be a challenge. Natural resources are well known as a source of several therapeutic agents for the treatment of neurologic disorders. Recently, chitosan (CTS) and its derivatives from arthropod exoskeletons, have attracted much attention as a drug delivery system to transport therapeutic substances across the BBB and thanks to other neuroprotective effects including the participation to the CNS regenerations scaffolds to replicate the extracellular matrix and microenvironment of the body. This review will discuss the place of natural resource therapy in targeting neurological disorders. In particular, it will highlight recent understanding and progress in the applications of CTS as drug delivery systems and their therapeutic effects on these disorders through tissue regeneration, as well as the molecular mechanisms by which they exert these effects.
Collapse
Affiliation(s)
- Youssef Ait Hamdan
- Interdisciplinary Laboratory of Research in Bio-Resources, Environment and Materials, Higher Normal School, Cadi Ayyad University, 40000 Marrakech, Morocco; Univ Rennes, CNRS, ISCR-UMR 6226, F-35000 Rennes, France.
| | - Bilal El-Mansoury
- Laboratory of Anthropogenic, Biotechnology and Health, Team physiopathology Nutritional, Neurosciences and Toxicology, Faculty of Sciences, Chouaib Doukkali University, Av. Des facultés, 24000 El Jadida, Morocco
| | - Samia Elouali
- Interdisciplinary Laboratory of Research in Bio-Resources, Environment and Materials, Higher Normal School, Cadi Ayyad University, 40000 Marrakech, Morocco; University of Mons (UMONS) - Laboratory of Polymeric and Composite Materials (LPCM), Center of Innovation and Research in Materials and Polymers (CIRMAP), Place du Parc 20, 7000 Mons, Belgium
| | - Khawla Rachmoune
- Interdisciplinary Laboratory of Research in Bio-Resources, Environment and Materials, Higher Normal School, Cadi Ayyad University, 40000 Marrakech, Morocco; Biotechnology and Biomolecule Engineering Unit, CNESTEN, Rabat, Morocco
| | - Anass Belbachir
- Center for Regenerative Medicine, CHU MOHAMMED VI, Marrakech, Morocco; Faculty of Medicine and Pharmacy, Cadi Ayyad University, Marrakech, Morocco
| | | | - Mohammed Rhazi
- Interdisciplinary Laboratory of Research in Bio-Resources, Environment and Materials, Higher Normal School, Cadi Ayyad University, 40000 Marrakech, Morocco
| |
Collapse
|
2
|
Hakami A, Narasimhan K, Comini G, Thiele J, Werner C, Dowd E, Newland B. Cryogel microcarriers for sustained local delivery of growth factors to the brain. J Control Release 2024; 369:404-419. [PMID: 38508528 DOI: 10.1016/j.jconrel.2024.03.023] [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: 12/15/2023] [Revised: 03/05/2024] [Accepted: 03/14/2024] [Indexed: 03/22/2024]
Abstract
Neurotrophic growth factors such as glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) have been considered as potential therapeutic candidates for neurodegenerative disorders due to their important role in modulating the growth and survival of neurons. However, clinical translation remains elusive, as their large size hinders translocation across the blood-brain barrier (BBB), and their short half-life in vivo necessitates repeated administrations. Local delivery to the brain offers a potential route to the target site but requires a suitable drug-delivery system capable of releasing these proteins in a controlled and sustained manner. Herein, we develop a cryogel microcarrier delivery system which takes advantage of the heparin-binding properties of GDNF and BDNF, to reversibly bind/release these growth factors via electrostatic interactions. Droplet microfluidics and subzero temperature polymerization was used to create monodisperse cryogels with varying degrees of negative charge and an average diameter of 20 μm. By tailoring the inclusion of 3-sulfopropyl acrylate (SPA) as a negatively charged moiety, the release duration of these two growth factors could be adjusted to range from weeks to half a year. 80% SPA cryogels and 20% SPA cryogels were selected to load GDNF and BDNF respectively, for the subsequent biological studies. Cell culture studies demonstrated that these cryogel microcarriers were cytocompatible with neuronal and microglial cell lines, as well as primary neural cultures. Furthermore, in vivo studies confirmed their biocompatibility after administration into the brain, as well as their ability to deliver, retain and release GDNF and BDNF in the striatum. Overall, this study highlights the potential of using cryogel microcarriers for long-term delivery of neurotrophic growth factors to the brain for neurodegenerative disorder therapeutics.
Collapse
Affiliation(s)
- Abrar Hakami
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK; Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Kaushik Narasimhan
- Pharmacology & Therapeutics and Galway Neuroscience Centre, University of Galway, H91 W5P7 Galway, Ireland
| | - Giulia Comini
- Pharmacology & Therapeutics and Galway Neuroscience Centre, University of Galway, H91 W5P7 Galway, Ireland
| | - Julian Thiele
- Leibniz-Institut für Polymerforschung Dresden e.V., 01069 Dresden, Germany; Institute of Chemistry, Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - Carsten Werner
- Leibniz-Institut für Polymerforschung Dresden e.V., 01069 Dresden, Germany
| | - Eilís Dowd
- Pharmacology & Therapeutics and Galway Neuroscience Centre, University of Galway, H91 W5P7 Galway, Ireland.
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK.
| |
Collapse
|
3
|
Buczyńska A, Sidorkiewicz I, Niemira M, Krętowski AJ, Węgrzyn P, Kosiński P, Zbucka-Krętowska M. Identification of MicroRNA Profiles in Fetal Spina Bifida: The Role in Pathomechanism and Diagnostic Significance. Int J Mol Sci 2024; 25:2896. [PMID: 38474143 DOI: 10.3390/ijms25052896] [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: 01/19/2024] [Revised: 02/16/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Distinct miRNA expression patterns may reflect anomalies related to fetal congenital malformations such as spinal bifida (SB). The aim of this preliminary study was to determine the maternal miRNA expression profile of women carrying fetuses with SB. Therefore, six women carrying fetuses with SB and twenty women with euploid healthy fetuses were enrolled in this study. Using NanoString technology, we evaluated the expression level of 798 miRNAs in both plasma and amniotic fluid samples. A downregulation of miR-1253, miR-1290, miR-194-5p, miR-302d-3p, miR-3144-3p, miR-4536-5p, miR-548aa + miR-548t-3p, miR-548ar-5p, miR-548n, miR-590-5p, miR-612, miR-627-5p, miR-644a, and miR-122-5p, and an upregulation of miR-320e, let-7b-5p, miR-23a-3p, miR-873-3p, and miR-30d-5p were identified in maternal amniotic fluid samples in SB when compared to the control group. The target genes of these miRNAs play a predominant role in regulating the synthesis of several biological compounds related to signaling pathways such as those regulating the pluripotency of stem cells. Moreover, the maternal plasma expression of miR-320e was increased in pregnancies with SB, and this marker could serve as a valuable non-invasive screening tool. Our results highlight the SB-specific miRNA signature and the differentially expressed miRNAs that may be involved in SB pathogenesis. Our findings emphasize the role of miRNA as a predictive factor that could potentially be useful in prenatal genetic screening for SB.
Collapse
Affiliation(s)
- Angelika Buczyńska
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Iwona Sidorkiewicz
- Clinical Research Support Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Magdalena Niemira
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Adam Jacek Krętowski
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Piotr Węgrzyn
- Department of Obstetrics, Perinatology and Gynecology, Medical University of Warsaw, 63A Zwirki i Wigury, 02-091 Warsaw, Poland
| | - Przemysław Kosiński
- Department of Obstetrics, Perinatology and Gynecology, Medical University of Warsaw, 63A Zwirki i Wigury, 02-091 Warsaw, Poland
| | - Monika Zbucka-Krętowska
- Department of Gynecological Endocrinology and Adolescent Gynecology, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| |
Collapse
|
4
|
Wang X, Ye L, Zhang K, Gao L, Xiao J, Zhang Y. Small Extracellular Vesicles Released from miR-211-5p-Overexpressed Bone Marrow Mesenchymal Stem Cells Ameliorate Spinal Cord Injuries in Rats. eNeuro 2024; 11:ENEURO.0361-23.2023. [PMID: 38351058 PMCID: PMC10866331 DOI: 10.1523/eneuro.0361-23.2023] [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: 09/18/2023] [Revised: 12/17/2023] [Accepted: 12/20/2023] [Indexed: 02/16/2024] Open
Abstract
Spinal cord injury (SCI) has become one of the common and serious diseases affecting patients' motor functions. The small extracellular vesicles secreted by bone marrow mesenchymal stem cells (BMSCs) have shown a promising prospect for the treatment of neurological diseases. BMSCs were collected from rat bones. Osteogenic and adipogenic differentiation of BMSCs was further determined. Small extracellular vesicles were obtained by high-speed centrifugation. Dual-luciferase reporter assay was performed to demonstrate the targeting of miR-211-5p to the cyclooxygenase 2 (COX2) mRNA. qRT-PCR and Western blot assay were used for the detection of the mRNA and protein expression. ELISA was performed to estimate the levels of proinflammatory factors in spinal cord tissues. Our results showed that miR-211-5p targeted COX2 mRNA and regulated the protein expression of COX2 in BMSCs. Extracellular vesicles released from miR-211-5p-overexpressed BMSCs ameliorated SCI-induced motor dysfunction and motor evoked potential impairments. Extracellular vesicles released from miR-211-5p-overexpressed BMSCs ameliorated SCI-induced COX2 expression and related inflammatory responses. In conclusion, small extracellular vesicles released from miR-211-5p-overexpressed BMSCs ameliorate spinal cord injuries in rats.
Collapse
Affiliation(s)
- Xianxiang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Lei Ye
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Ke Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Lu Gao
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Jin Xiao
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Yiquan Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| |
Collapse
|
5
|
Wang Y, Zhan Y, Ji C, Shi C, Han J. Houttuynia cordata Thunb repairs steroid-induced avascular necrosis of the femoral head through regulating NF-κB signaling pathway. Toxicon 2023; 233:107270. [PMID: 37652100 DOI: 10.1016/j.toxicon.2023.107270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/13/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
To investigate the influences of Houttuynia cordata Thunb (HCT) in steroid-induced avascular necrosis of the femoral head (SANFH), we conducted a comprehensive study evaluating the effects of HCT on various aspects. Cell Counting Kit-8 assay was used to examine bone marrow stem cells (BMSCs) cell viability. Flow cytometry and lactate dehydrogenase detection assay were conducted to determine cell apoptosis. The levels of apoptosis-related proteins, osteogenic-related markers, inflammatory factors, and nuclear factor kappa B (NF-κB) pathway-associated proteins were determined via western blotting. Hematoxylin and eosin and terminal deoxynucleotidyl transferase dUTP nick-end labeling assays were utilized to verify the effects of HCT in SANFH rats. Our findings revealed that HCT could enhanced cell viability and arrested cell apoptosis in dexamethasone (Dex)-treated BMSCs. Dex increased the levels of cleaved caspase-3, Bcl2-associated X, interleukin (IL)-1β, IL-18, IL-6, p65, and inhibitor of NF-κB kinase β (IKKβ), while this promoting trend was weakened by HCT. Moreover, pyrrolidine dithiocarbamate (PDTC, an inhibitor of NF-κB signaling pathway) further increased the inhibitory role of apoptosis and the levels of IL-1β, IL-18, and IL-6 and the promotional effect of the levels of RUNX2 and ALP in Dex-treated BMSCs. The in-vivo assays showed that HCT decreased the percentage of empty lacunae, apoptosis, and the levels of IL-1β, IL-18, IL-6, p65, and IKKβ in SANFH rats. In conclusion, our study demonstrated that HCT relieved SANFH, which might be possibly achieved by NF-κB pathway.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Medical Insurance Office, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, Shandong, 266000, PR China
| | - Yaqi Zhan
- Hematology Department, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, Shandong, 266000, PR China
| | - Chunxiao Ji
- Hematology Department, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, Shandong, 266000, PR China
| | - Chunlei Shi
- Hematology Department, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, Shandong, 266000, PR China
| | - Jie Han
- Hematology Department, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, Shandong, 266000, PR China.
| |
Collapse
|
6
|
Nguyen LT, Le HT, Nguyen KT, Bui HT, Nguyen APT, Ngo DV, Hoang DM, Ngo MD. Outcomes of autologous bone marrow mononuclear cell administration in the treatment of neurologic sequelae in children with spina bifida. Stem Cell Res Ther 2023; 14:115. [PMID: 37118832 PMCID: PMC10148418 DOI: 10.1186/s13287-023-03349-w] [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: 08/02/2022] [Accepted: 04/19/2023] [Indexed: 04/30/2023] Open
Abstract
BACKGROUND To evaluate the safety and efficacy of autologous bone marrow mononuclear cell (BMMNC) infusion in the management of neurological sequelae in children with spina bifida (SB). METHODS BMMNCs were harvested from bilateral anterior iliac crests. Two intrathecal BMMNC administrations were performed with an interval of 6 months. The measurements of outcomes included clinical assessments, cystomanometry and rectomanometry. RESULTS Eleven children with SB underwent autologous BMMNC infusions from 2016 to 2020. There were no severe adverse events during the study period. The number of patients requiring assistance to expel stools decreased from 11 before cell infusion to 3 after the second cell infusion. The number of patients who had urine leakage decreased from 9 patients at baseline to 3 patients after the second BMMNC infusion. The mean bladder capacity increased from 127.7 ± 59.2 ml at baseline to 136.3 ± 54.8 ml at six months and to 158.3 ± 56.2 ml at 12 months after BMMNC infusions. Detrusor pressure (pdet) decreased from 32.4 ± 22.0 cm H2O at baseline to 21.9 ± 11.8 cm H2O after 12 months of follow-up. At baseline, six patients could walk independently. After the 2nd infusion, eight patients could walk independently. CONCLUSION Intrathecal infusions of autologous bone marrow mononuclear cells are safe and may improve bowel, bladder, and motor function in children with SB. TRIAL REGISTRATION NCT, NCT05472428. Registered July 25, 2022- Retrospectively registered, https://www. CLINICALTRIALS gov/ct2/show/NCT05472428 .
Collapse
Affiliation(s)
- Liem Thanh Nguyen
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, 458 Minh Khai, Hanoi, Vietnam.
- College of Health Science, VinUniversity, Vinhomes Ocean Park, Gia Lam District, Hanoi, Vietnam.
- Vinmec International Hospital - Times City, Vinmec Health Care System, 458 Minh Khai, Hanoi, Vietnam.
| | - Huong Thu Le
- Vinmec International Hospital - Times City, Vinmec Health Care System, 458 Minh Khai, Hanoi, Vietnam
| | - Kien Trung Nguyen
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, 458 Minh Khai, Hanoi, Vietnam
| | - Hang Thi Bui
- Vinmec International Hospital - Times City, Vinmec Health Care System, 458 Minh Khai, Hanoi, Vietnam
| | - Anh Phuong Thi Nguyen
- Vinmec International Hospital - Times City, Vinmec Health Care System, 458 Minh Khai, Hanoi, Vietnam
| | - Doan Van Ngo
- Vinmec International Hospital - Times City, Vinmec Health Care System, 458 Minh Khai, Hanoi, Vietnam
| | - Duc Minh Hoang
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, 458 Minh Khai, Hanoi, Vietnam
| | - Minh Duy Ngo
- Vinmec International Hospital - Times City, Vinmec Health Care System, 458 Minh Khai, Hanoi, Vietnam
| |
Collapse
|
7
|
Damianos A, Sammour I. Barriers in translating stem cell therapies for neonatal diseases. Semin Perinatol 2023; 47:151731. [PMID: 36990922 DOI: 10.1016/j.semperi.2023.151731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Over the last 20 years, stem cells of varying origin and their associated secretome have been investigated as a therapeutic option for a myriad of neonatal models of disease, with very promising results. Despite the devastating nature of some of these disorders, translation of the preclinical evidence to the bedside has been slow. In this review, we explore the existing clinical evidence for stem cell therapies in neonates, highlight the barriers faced by researchers and suggest potential solutions to move the field forward.
Collapse
Affiliation(s)
- Andreas Damianos
- Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, Ohio
| | - Ibrahim Sammour
- Riley Hospital for Children, Indiana University, Indianapolis, USA.
| |
Collapse
|
8
|
Leukemia Inhibitory Factor Facilitates Self-Renewal and Differentiation and Attenuates Oxidative Stress of BMSCs by Activating PI3K/AKT Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5772509. [PMID: 36105481 PMCID: PMC9467750 DOI: 10.1155/2022/5772509] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/09/2022] [Accepted: 07/15/2022] [Indexed: 11/17/2022]
Abstract
Objective. Transplantation of bone marrow-derived mesenchymal stem cells (BMSCs) remains a hopeful therapeutic approach for bone defect reconstruction. Herein, we investigated the effects and mechanisms of leukemia inhibitory factor (LIF) in the function and viability of hypoxic BMSCs as well as bone defect repair. Methods. The effects of LIF on apoptosis (flow cytometry, TUNEL staining), mitochondrial activity (JC-1 staining), proliferation (colony formation, EdU staining), and differentiation (CD105, CD90, and CD29 via flow sorting) were examined in hypoxic BMSCs. LIF, LIFR, gp130, Keap1, Nrf2, antioxidant enzymes (SOD1, catalase, GPx-3), bone-specific matrix proteins (ALP, BSP, OCN), PI3K, and Akt were detected via immunoblotting or immunofluorescent staining. BMSCs combined with biphasic calcium phosphate scaffolds were implanted into calvarial bone defect mice, and the therapeutic effect of LIF on bone defect was investigated. Results. Hypoxic BMSCs had increased apoptosis and oxidative stress and reduced mitochondrial activity. Additionally, LIF, LIFR, and gp130 were upregulated and PI3K/Akt activity was depressed in hypoxic BMSCs. Upregulated LIF alleviated apoptosis and oxidative stress and heightened mitochondrial activity and PI3K/Akt signaling in hypoxic BMSCs. Additionally, LIF overexpression promoted self-renewal and osteogenic differentiation of BMSCs with hypoxic condition. Mechanically, LIF facilitated self-renewal and differentiation as well as attenuated oxidative stress of BMSCs through enhancing PI3K/AKT signaling activity. Implantation of LIF-overexpressed BMSC-loaded BCP scaffolds promoted osteogenesis as well as alleviated oxidative stress and apoptosis through PI3K/Akt signaling. Conclusion. Our findings demonstrate that LIF facilitates self-renewal and differentiation and attenuates oxidative stress of BMSCs by PI3K/AKT signaling.
Collapse
|
9
|
Uy MNAR, Tantengco OAG. Investigating the landscape and trajectory of spina bifida research in Asia: a bibliometric analysis. Childs Nerv Syst 2022; 38:1581-1591. [PMID: 35474541 DOI: 10.1007/s00381-022-05527-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/10/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Spina bifida is a type of a neural tube defect which affects 243.14 per 100,000 babies in Asia. Research articles on spina bifida have increased in the recent years. However, no study has focused on the research trends in this field in Asia. METHODS A systematic review of literature on spina bifida in Asia was performed using the Scopus database from inception to 2020. All published studies on spina bifida conducted in or published by authors from Asia were included in our analysis. Bibliometric information was obtained from Scopus and bibliometrics diagrams were created using VOSviewer software. RESULTS A total of 652 articles were obtained in this study. The number of publications showed an upward trend starting 2000s. The country with the greatest number of publications was Japan while All India Institute of Medical Sciences was the most productive institution in spina bifida research in Asia. The current focus of this field in Asia was prevalence of spina bifida, prenatal diagnosis, folic acid supplementation, and complications of spina bifida. Future areas of research in spina bifida include the genetic basis of neural tube defects and the use of stem cell technology as therapies for spina bifida. CONCLUSION This is the first bibliometric analysis on spina bifida in Asia. It showed the trend and future areas of research on spina bifida in Asia. Despite the increase in scientific literature on spina bifida research, more research outputs and collaborations are needed especially in developing countries in Asia.
Collapse
Affiliation(s)
| | - Ourlad Alzeus G Tantengco
- College of Medicine, University of the Philippines Manila, Pedro Gil Street, Ermita, Manila, Philippines.
| |
Collapse
|
10
|
Advances in Regulatory Strategies of Differentiating Stem Cells towards Keratocytes. Stem Cells Int 2022; 2022:5403995. [PMID: 35140792 PMCID: PMC8820938 DOI: 10.1155/2022/5403995] [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: 07/20/2021] [Revised: 12/16/2021] [Accepted: 01/05/2022] [Indexed: 11/17/2022] Open
Abstract
Corneal injury is a commonly encountered clinical problem which led to vision loss and impairment that affects millions of people worldwide. Currently, the available treatment in clinical practice is corneal transplantation, which is limited by the accessibility of donors. Corneal tissue engineering appears to be a promising alternative for corneal repair. However, current experimental strategies of corneal tissue engineering are insufficient due to inadequate differentiation of stem cell into keratocytes and thus cannot be applied in clinical practice. In this review, we aim to clarify the role and effectiveness of both biochemical factors, physical regulation, and the combination of both to induce stem cells to differentiate into keratocytes. We will also propose novel perspectives of differentiation strategy that may help to improve the efficiency of corneal tissue engineering.
Collapse
|
11
|
Ma L, Wei X, Ma W, Liu Y, Wang Y, He Y, Jia S, Wang Y, Luo W, Liu D, Huang T, Yan J, Gu H, Bai Y, Yuan Z. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:539-551. [PMID: 35325230 PMCID: PMC9154334 DOI: 10.1093/stcltm/szac009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 01/24/2022] [Indexed: 11/12/2022] Open
Abstract
Spinal bifida aperta (SBA) is a congenital malformation with a high incidence. Bone marrow mesenchymal stem cell (BMSC) transplantation has the potential to repair the structure of damaged tissues and restore their functions. This is an optional treatment that can be used as a supplement to surgery in the treatment of SBA. However, the application of BMSCs is limited, as the neuronal differentiation rate of BMSCs is not satisfactory when used in treating severe SBA. Thus, we aimed to assess the effect of neural stem cell (NSC)-derived exosomes on BMSC neuronal differentiation and observe the therapeutic effect in an ex vivo rat SBA embryo model. We found that NSC-derived exosomes increased the neuronal differentiation rate of BMSCs in vitro and in the SBA embryo model ex vivo. Proteomic analysis showed that NSC-derived exosomes were enriched in Netrin1, which positively regulated neuronal differentiation. Netrin1 increased the neuronal differentiation rate of BMSCs and NSCs and upregulated the expression of the neuronal markers, microtubule-associated protein (Map2), neurofilament, and β3-tubulin. Bioinformatic analysis revealed that Netrin1 treatment increased the expression of the transcription factors Hand2 and Phox2b, related to neuronal differentiation. Furthermore, the Netrin1-induced NSC neuronal differentiation was significantly blocked by Phox2b knockdown. We suggest that NSC-derived exosomal Netrin1 induces neuronal differentiation via the Hand2/Phox2b axis by upregulating the expression of Hand2 and Phox2b. Therefore, NSC-derived exosomes are a critical inducer of BMSC neuronal differentiation and represent a potential treatment agent that can benefit BMSC treatment in SBA.
Collapse
Affiliation(s)
- Ling Ma
- Key laboratory of Health Ministry for Congenital Malformation, Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning, People’s Republic of China
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Xiaowei Wei
- Key laboratory of Health Ministry for Congenital Malformation, Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Wei Ma
- Key laboratory of Health Ministry for Congenital Malformation, Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Yusi Liu
- Key laboratory of Health Ministry for Congenital Malformation, Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Yanfu Wang
- Key laboratory of Health Ministry for Congenital Malformation, Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Yiwen He
- Key laboratory of Health Ministry for Congenital Malformation, Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Shanshan Jia
- Key laboratory of Health Ministry for Congenital Malformation, Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Yu Wang
- Key laboratory of Health Ministry for Congenital Malformation, Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning, People’s Republic of China
- Department of Ultrasound, Shengjing Hospital, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Wenting Luo
- Key laboratory of Health Ministry for Congenital Malformation, Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Dan Liu
- Key laboratory of Health Ministry for Congenital Malformation, Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Tianchu Huang
- Key laboratory of Health Ministry for Congenital Malformation, Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Jiayu Yan
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Hui Gu
- Key laboratory of Health Ministry for Congenital Malformation, Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Yuzuo Bai
- Key laboratory of Health Ministry for Congenital Malformation, Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Zhengwei Yuan
- Key laboratory of Health Ministry for Congenital Malformation, Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning, People’s Republic of China
- Corresponding author: Zhengwei Yuan, Key laboratory of Health Ministry for Congenital Malformation, Department of Pediatric Surgery, Shengjing Hospital, China Medical University, No. 36, Sanhao Street, Heping District, Shenyang 110004, China. Tel: +86 24 23929903;
| |
Collapse
|
12
|
Li Y, Wang P, Hu XD, Zeng JD, Fang C, Gan Y, Peng FY, Yang XL, Luo D, Li B, Su S. Implantation of Bone Marrow Mesenchymal Stem Cells into Small Intestinal Submucosa Improves Bile Duct Injury in Rabbits. Tissue Eng Regen Med 2021; 18:887-893. [PMID: 34216376 DOI: 10.1007/s13770-021-00351-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 04/30/2021] [Accepted: 05/08/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Bile duct injury (BDI), which may occur during cholecystectomy procedures and living-donor liver transplantation, leads to life-altering complications and significantly increased mortality and morbidity. Tissue engineering, as an emerging method, has shown great potential to treat BDI. Here, we aimed to explore the application of small intestinal submucosa (SIS) matrix composites with bone marrow mesenchymal stem cells (BMSCs) to treat BDI in a rabbit model. METHODS Rabbit-derived BMSCs were used as seed cells. Porcine SIS was used as the support material. Five centimetres of the common bile duct was dissected, and 1/3-1/2 of the anterior wall diameter was transversely incised to construct the rabbit BDI model. Then, SIS materials without/with BMSCs were inserted into the common bile duct of the BDI rabbits. After 1, 2, 4, and 8 weeks of implantation, the common bile duct was removed. Haematoxylin and eosin (HE) staining was used to assess pathological alterations in the common bile duct, while immunohistochemical staining and western blotting were used to detect expression of the epithelial cell markers CK19 and E-cadherin. Scanning electron microscopy was used to evaluate BMSC growth. RESULTS Compared with BMSCs alone, SIS-attached BMSCs had increased growth. HE staining showed that the injured bile duct healed well and that the complex gradually degraded as the time from implantation increased. Immunohistochemical staining and western blotting showed that compared with the control group, the in vivo complex group had significantly elevated expression levels of CK19 and E-cadherin. CONCLUSION BMSC implantation into SIS could improve BDI in rabbits, which might have clinical value for BDI treatment.
Collapse
Affiliation(s)
- Ying Li
- Department of Plastic and Burns Surgery, The Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Luzhou, 646000, Sichuan, People's Republic of China
| | - Piao Wang
- Department of General Surgery (Hepatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Xiao-Dong Hu
- Department of General Surgery (Hepatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Jing-da Zeng
- Department of Orthopaedic, Hospital of Traditional Chinese Medicine, affiliated to Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Cheng Fang
- Department of General Surgery (Hepatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Yu Gan
- Department of General Surgery (Hepatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Fang-Yi Peng
- Department of General Surgery (Hepatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Xiao-Li Yang
- Department of General Surgery (Hepatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - De Luo
- Department of General Surgery (Hepatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Bo Li
- Department of General Surgery (Hepatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China. .,Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, China.
| | - Song Su
- Department of General Surgery (Hepatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China. .,Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, China.
| |
Collapse
|
13
|
Liang Y, Zhou R, Liu X, Liu Z, You L, Chen C, Ye X. Investigation into the effects of leukemia inhibitory factor on the bone repair capacity of BMSCs-loaded BCP scaffolds in the mouse calvarial bone defect model. J Bioenerg Biomembr 2021; 53:381-391. [PMID: 34110599 DOI: 10.1007/s10863-021-09899-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/19/2021] [Indexed: 01/09/2023]
Abstract
Leukemia inhibitory factor (LIF) is known to play a major role in bone physiology. In the present study, we examined the in vitro effects of LIF on osteoblast differentiation of bone marrow stem cells (BMSCs) and explored in vivo effects of LIF on the bone repair capacity of BMSCs-loaded biphasic calcium phosphate (BCP) scaffolds in mouse calvarial bone defect model. The mRNA and protein expression levels in the BMSCs were determined by quantitative real-time PCR and western blot, respectively; the in vitro osteoblast differentiation of the BMSCs was evaluated by using Alizarin Red S staining. The bone volume and bone density in the repaired calvarial bone defect were determined by Micro-CT. Bone regeneration was also histologically evaluated by hematoxylin and eosin staining and Masson's trichrome staining. Hypoxia treatment induced the up-regulation of Lif mRNA and LIF protein in the BMSCs. Lif overexpression up-regulated the mRNA expression levels of osteopontin and Runt-related transcription factor 2, and increased intensity of Alizarin Red S staining in the BMSCs; while Lif silence exerted the opposite effects. The in vivo studies showed that implantation of Lif-overexpressing BMSCs-loaded BCP scaffolds significantly increased the bone volume and bone density at 4 and 8 weeks after transplantation, and promoted the regeneration of bone tissues in the mouse calvarial bone defect at 8 weeks after transplantation when compared to the BMSCs-loaded BCP scaffolds group; while Lif-silencing BMSCs-loaded BCP scaffolds had the opposite effects. The present study for the first time demonstrated that LIF promoted the in vitro osteoblast differentiation of hypoxia-treated BMSCs; and further studies revealed that LIF exerted enhanced effects on the bone repair capacity of BMSCs-load BCP scaffolds in mouse calvarial bone defect model. However, future studies are warranted to determine the detailed mechanisms of LIF in the large-scale bone defect repair.
Collapse
Affiliation(s)
- Youde Liang
- Department of Stomatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China. .,Department of Stomatology, Southern University of Science and Technology Yantian Hospital, Shenzhen, China.
| | - Ruiping Zhou
- Department of Stomatology, Southern University of Science and Technology Yantian Hospital, Shenzhen, China
| | - Xin Liu
- Department of Stomatology, Southern University of Science and Technology Yantian Hospital, Shenzhen, China
| | - Zhikang Liu
- Department of Stomatology, Southern University of Science and Technology Yantian Hospital, Shenzhen, China
| | - Lin You
- Department of Stomatology, Southern University of Science and Technology Yantian Hospital, Shenzhen, China
| | - Chang Chen
- Department of Stomatology, Southern University of Science and Technology Yantian Hospital, Shenzhen, China
| | - Xiaoling Ye
- Department of Stomatology, Southern University of Science and Technology Yantian Hospital, Shenzhen, China
| |
Collapse
|
14
|
Xu L, Gao W, Bai S, Duan H, Pan X, Wu W. MEF/KSF-conditioned culture medium: An effective method for in vitro culture of mouse dermal papilla cells with osteogenic differentiation potential. Exp Ther Med 2021; 22:828. [PMID: 34149874 PMCID: PMC8200806 DOI: 10.3892/etm.2021.10260] [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: 01/15/2020] [Accepted: 01/08/2021] [Indexed: 11/06/2022] Open
Abstract
Hair follicle stem cells are pluripotent and have a self-renewal capacity and multi-differentiation potential in vitro. As hair follicle stem cells can be easily sampled from the skin and hair of clinical patients at a considerable quantity, these cells have potential applications in wound repair and skin tissue engineering. Effective approaches for the in vitro culture and amplification of mouse hair follicle stem cells, as well as the in vitro osteogenic differentiation potential and cell source when obtaining mouse-separated cells were examined. Serial subculture was performed in different culture systems. Cell source was detected based on the relevant surface markers derived from mouse hair follicles at the gene and protein levels, and the differential potential was determined. The proliferative ability of hair follicle-derived stem cells obtained from mouse embryonic fibroblast (MEF)/keratinocyte serum-free medium (KSF)-conditioned medium was the highest among all culture systems. The induced group had a stronger osteogenic differentiation potential compared with the non-induced group, indicating that the cells obtained from MEF/KSF-conditioned medium were cells derived from the hair follicle dermal papilla. Therefore, the strong osteogenic differentiation potential of the hair follicle-derived mesenchymal stem cells was screened with MEF/KSF-conditioned culture medium following amplification, and biological characteristics similar to those of hair follicle dermal papilla cells were observed.
Collapse
Affiliation(s)
- Liang Xu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Wenlan Gao
- Department of Stomatology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Shanshan Bai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Huichuan Duan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Xiaogang Pan
- Department of Orthodontics, Shanghai Ninth Peoples' Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Wei Wu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
15
|
Shah SA, Sohail M, Khan SA, Kousar M. Improved drug delivery and accelerated diabetic wound healing by chondroitin sulfate grafted alginate-based thermoreversible hydrogels. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 126:112169. [PMID: 34082970 DOI: 10.1016/j.msec.2021.112169] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/20/2021] [Accepted: 05/03/2021] [Indexed: 12/16/2022]
Abstract
Injectable hydrogels with multifunctional tunable properties comprising biocompatibility, anti-oxidative, anti-bacterial, and/or anti-infection are highly preferred to efficiently promote diabetic wound repair and its development remains a challenge. In this study, we report chondroitin sulphate (CS) and sodium alginate (SA)-based injectable hydrogel using solvent casting method loaded with curcumin that could potentiate reepithelization, increase angiogenesis, and collagen deposition at wound microenvironment to endorse healing cascade. The physical interaction and self-assembly of chondroitin sulfate grafted alginate (CS-Alg-g-PF127) hydrogel were confirmed using nuclear magnetic resonance (1H NMR) and Fourier transformed infrared spectroscopy (FT-IR), and cytocompatibility was confirmed by fibroblast viability assay. The Masson's trichrome (MT) and hematoxylin and eosin (H&E) results revealed that blank chondroitin sulfate grafted alginate (CS-Alg-g-PF127) and CUR loaded CS-Alg-g-PF127 hydrogel had promising tissue regenerative ability, and showing enhanced wound healing compared to other treatment groups. The controlled release of CUR from injectable hydrogel was evaluated by drug release studies and pharmacokinetic profile (PK) using high-performance liquid chromatography (HPLC) that exhibited the mean residence time (MRT) and area under the curve (AUC) was increased up to 16.18 h and 203.64 ± 30.1 μg/mL*h, respectively. Cytotoxicity analysis of the injectable hydrogels using 3 T3-L1 fibroblasts cells and in vivo toxicity evaluated by subcutaneous injection for 24 h followed by histological examination, confirmed good biocompatibility of CUR loaded CS-Alg-g-PF127 hydrogel. Interestingly, the results of in vivo wound healing by injectable hydrogel showed the upregulation of fibroblasts-like cells, collagen deposition, and differentiated keratinocytes stimulating dermo-epidermal junction, which might endorse that they are potential candidates for excisional wound healing models.
Collapse
Affiliation(s)
- Syed Ahmed Shah
- Department of Pharmacy, COMSATS University, Islamabad, Abbottabad Campus, 22010, Pakistan
| | - Muhammad Sohail
- Department of Pharmacy, COMSATS University, Islamabad, Abbottabad Campus, 22010, Pakistan.
| | - Shujaat Ali Khan
- Department of Pharmacy, COMSATS University, Islamabad, Abbottabad Campus, 22010, Pakistan
| | - Mubeen Kousar
- Department of Pharmacy, COMSATS University, Islamabad, Abbottabad Campus, 22010, Pakistan
| |
Collapse
|
16
|
Thermosensitive quaternized chitosan hydrogel scaffolds promote neural differentiation in bone marrow mesenchymal stem cells and functional recovery in a rat spinal cord injury model. Cell Tissue Res 2021; 385:65-85. [PMID: 33760948 DOI: 10.1007/s00441-021-03430-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 01/26/2021] [Indexed: 12/24/2022]
Abstract
A thermosensitive quaternary ammonium chloride chitosan/β-glycerophosphate (HACC/β-GP) hydrogel scaffold combined with bone marrow mesenchymal stem cells (BMSCs) transfected with an adenovirus containing the glial cell-derived neurotrophic factor (GDNF) gene (Ad-rGDNF) was applied to spinal cord injury (SCI) repair. The BMSCs from rats were transfected with Ad-rGDNF, resulting in the expression of GDNF mRNA in the BMSCs increasing and their spontaneous differentiation into neural-like cells expressing neural markers such as NF-200 and GFAP. After incubation with HACC/β-GP hydrogel scaffolds for 2 weeks, neuronal differentiation of the BMSCs was confirmed using immunofluorescence (IF), and the expression of GDNF by the BMSCs was detected by Western blot at different time points. MTT assay and scanning electron microscopy confirmed that the HACC scaffold provides a non-cytotoxic microenvironment that supports cell adhesion and growth. Rats with SCI were treated with BMSCs, BMSCs carried by the HACC/β-GP hydrogel (HACC/BMSCs), Ad-rGDNF-BMSCs, or Ad-rGDNF-BMSCs carried by the hydrogel (HACC/GDNF-BMSCs). Animals were sacrificed at 2, 4, and 6 weeks of treatment. IF staining and Western blot were performed to detect the expression of NeuN, NF-200, GFAP, CS56, and Bax in the lesion sites of the injured spinal cord. Upon treatment with HACC/BMSCs, NF200 and GFAP were upregulated but CS56 and Bax were downregulated in the SCI lesion site. Furthermore, transplantation of HACC/GDNF-BMSCs into an SCI rat model significantly improved BBB scores and regeneration of the spinal cord. Thus, HACC/β-GP hydrogel scaffolds show promise for functional recovery in spinal cord injury patients.
Collapse
|
17
|
Soltani Khaboushan A, Shakibaei M, Kajbafzadeh AM, Majidi Zolbin M. Prenatal Neural Tube Anomalies: A Decade of Intrauterine Stem Cell Transplantation Using Advanced Tissue Engineering Methods. Stem Cell Rev Rep 2021; 18:752-767. [PMID: 33742349 DOI: 10.1007/s12015-021-10150-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2021] [Indexed: 10/21/2022]
Abstract
Neural tube defects (NTDs) are among the most common congenital defects during neurulation. Spina bifida is a type of NTD that can occur in different forms. Since myelomeningocele (MMC) is the most severe form of spina bifida, finding a satisfactory treatment for MMC is a gold standard for the treatment of spina bifida. The Management of Myelomeningocele Study (MOMS) demonstrated that intrauterine treatment of spina bifida could ameliorate the complications associated with spina bifida and would also reduce the placement of ventriculoperitoneal (VP) shunt by 50%. Recently developed tissue engineering (TE) approaches using scaffolds, stem cells, and growth factors allow treatment of the fetus with minimally invasive methods and promising outcomes. The application of novel patches with appropriate stem cells and growth factors leads to better coverage of the defect with fewer complications. These approaches with less invasive surgical procedures, even in animal models with similar characteristics as the human MMC defect, paves the way for the modern application of less invasive surgical methods. Significantly, the early detection of these problems and applying these approaches can increase the potential efficacy of MMC treatment with fewer complications. However, further studies should be conducted to find the most suitable scaffolds and stem cells, and their application should be evaluated in animal models. This review intends to discuss advanced TE methods for treating MMC and recent successes in increasing the efficacy of the treatment.
Collapse
Affiliation(s)
- Alireza Soltani Khaboushan
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences, No. 62, Dr. Gharib's Street, Keshavarz Boulevard, Tehran, 1419433151, Iran.,Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, Pettenkoferstrasse 11, D-80336, Munich, Germany
| | - Abdol-Mohammad Kajbafzadeh
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences, No. 62, Dr. Gharib's Street, Keshavarz Boulevard, Tehran, 1419433151, Iran.
| | - Masoumeh Majidi Zolbin
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences, No. 62, Dr. Gharib's Street, Keshavarz Boulevard, Tehran, 1419433151, Iran.
| |
Collapse
|
18
|
Eigel D, Werner C, Newland B. Cryogel biomaterials for neuroscience applications. Neurochem Int 2021; 147:105012. [PMID: 33731275 DOI: 10.1016/j.neuint.2021.105012] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/16/2022]
Abstract
Biomaterials in the form of 3D polymeric scaffolds have been used to create structurally and functionally biomimetic constructs of nervous system tissue. Such constructs can be used to model defects and disease or can be used to supplement neuronal tissue regeneration and repair. One such group of biomaterial scaffolds are hydrogels, which have been widely investigated for cell/tissue culture and as cell or molecule delivery systems in the field of neurosciences. However, a subset of hydrogels called cryogels, have shown to possess several distinct structural advantages over conventional hydrogel networks. Their macroporous structure, created via the time and resource efficient fabrication process (cryogelation) not only allows mass fluid transport throughout the structure, but also creates a high surface area to volume ratio for cell growth or drug loading. In addition, the macroporous structure of cryogels is ideal for applications in the central nervous system as they are very soft and spongey, yet also robust, which makes them a user-friendly and reproducible tool to address neuroscience challenges. In this review, we aim to provide the neuroscience community, who may not be familiar with the fundamental concepts of cryogels, an accessible summary of the basic information that pertain to their use in the brain and nervous tissue. We hope that this review shall initiate creative ways that cryogels could be further adapted and employed to tackle unsolved neuroscience challenges.
Collapse
Affiliation(s)
- Dimitri Eigel
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Str. 6, 01069, Dresden, Germany
| | - Carsten Werner
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Str. 6, 01069, Dresden, Germany; Technische Universität Dresden, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Ben Newland
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Str. 6, 01069, Dresden, Germany; School of Pharmacy and Pharmaceutical Sciences, Cardiff University, CF10 3NB, Cardiff, Wales, UK.
| |
Collapse
|
19
|
Kunpalin Y, Subramaniam S, Perin S, Gerli MFM, Bosteels J, Ourselin S, Deprest J, De Coppi P, David AL. Preclinical stem cell therapy in fetuses with myelomeningocele: A systematic review and meta-analysis. Prenat Diagn 2021; 41:283-300. [PMID: 33427329 PMCID: PMC7611444 DOI: 10.1002/pd.5887] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/16/2020] [Accepted: 12/15/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVE We performed a systematic review to summarize the efficacy and safety of in utero stem cells application in preclinical models with myelomeningocele (MMC). METHODS The study was registered with PROSPERO (CRD42019160399). We searched MEDLINE, Embase, Web of Science, Scopus and CENTRAL for publications articles on stem cell therapy in animal fetuses with MMC until May 2020. Publication quality was assessed by the SYRCLE's tool. Meta-analyses were pooled if studies were done in the same animal model providing similar type of stem cell used and outcome measurements. Narrative synthesis was performed for studies that could not be pooled. RESULTS Nineteen and seven studies were included in narrative and quantitative syntheses, respectively. Most used mesenchymal stem cells (MSCs) and primarily involved ovine and rodent models. Both intra-amniotic injection of allogeneic amniotic fluid (AF)-MSCs in rat MMC model and the application of human placental (P)-MSCs to the spinal cord during fetal surgery in MMC ovine model did not compromise fetal survival rates at term (rat model, relative risk [RR] 1.03, 95% CI 0.92-1.16; ovine model, RR 0.94, 95% CI 0.78-1.13). A single intra-amniotic injection of allogeneic AF-MSCs into rat MMC model was associated with a higher rate of complete defect coverage compared to saline injection (RR 16.35, 95% CI 3.27-81.79). The incorporation of human P-MSCs as a therapeutic adjunct to fetal surgery in the ovine MMC model significantly improved sheep locomotor rating scale after birth (mean difference 5.18, 95% CI 3.36-6.99). CONCLUSIONS Stem cell application during prenatal period in preclinical animal models is safe and effective.
Collapse
Affiliation(s)
- Yada Kunpalin
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, UK.,Department of Development and Regeneration, Cluster Woman and Child, Biomedical Sciences, KU Leuven, Leuven, Belgium
| | - Sindhu Subramaniam
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Silvia Perin
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Mattia F M Gerli
- Great Ormond Street Institute of Child Health, University College London, London, UK.,Division of Surgery and Interventional Science, Royal Free Hospital, University College London, London, UK
| | - Jan Bosteels
- Department of Development and Regeneration, Cluster Woman and Child, Biomedical Sciences, KU Leuven, Leuven, Belgium.,Cochrane Belgium, Belgian Centre for Evidence-Based Medicine (Cebam), Leuven, Belgium
| | - Sebastien Ourselin
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| | - Jan Deprest
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, UK.,Department of Development and Regeneration, Cluster Woman and Child, Biomedical Sciences, KU Leuven, Leuven, Belgium.,Department of Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium
| | - Paolo De Coppi
- Department of Development and Regeneration, Cluster Woman and Child, Biomedical Sciences, KU Leuven, Leuven, Belgium.,Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Anna L David
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, UK.,Department of Development and Regeneration, Cluster Woman and Child, Biomedical Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
20
|
Wei X, Ma W, Gu H, Liu D, Luo W, Bai Y, Wang W, Lui VCH, Yang P, Yuan Z. Transamniotic mesenchymal stem cell therapy for neural tube defects preserves neural function through lesion-specific engraftment and regeneration. Cell Death Dis 2020; 11:523. [PMID: 32655141 PMCID: PMC7354991 DOI: 10.1038/s41419-020-2734-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 12/24/2022]
Abstract
Neural tube defects (NTDs) lead to prenatal mortality and lifelong morbidity. Currently, surgical closure of NTD lesions results in limited functional recovery. We previously suggested that nerve regeneration was critical for NTD therapy. Here, we report that transamniotic bone marrow-derived mesenchymal stem cell (BMSC) therapy for NTDs during early development may achieve beneficial functional recovery. In our ex vivo rat embryonic NTD model, BMSCs injected into the amniotic cavity spontaneously migrated into the defective neural tissue. Hepatocyte growth factor and its receptor c-MET were found to play critical roles in this NTD lesion-specific migration. Using the in vivo rat fetal NTD model, we further discovered that the engrafted BMSCs specifically differentiated into the cell types of the defective tissue, including skin and different types of neurons in situ. BMSC treatment triggered skin repair in fetuses, leading to a 29.9 ± 5.6% reduction in the skin lesion area. The electrophysiological functional recovery assay revealed a decreased latency and increased motor-evoked potential amplitude in the BMSC-treated fetuses. Based on these positive outcomes, ease of operation, and reduced trauma to the mother and fetus, we propose that transamniotic BMSC administration could be a new effective therapy for NTDs.
Collapse
Affiliation(s)
- Xiaowei Wei
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Wei Ma
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Hui Gu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Dan Liu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Wenting Luo
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Yuzuo Bai
- Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, PR China
| | - Weilin Wang
- Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, PR China
| | - Vincent Chi Hang Lui
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, China
| | - Peixin Yang
- Departments of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Zhengwei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
21
|
Cell therapy for prenatal repair of myelomeningocele: A systematic review. Curr Res Transl Med 2020; 68:183-189. [PMID: 32624428 DOI: 10.1016/j.retram.2020.04.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/13/2020] [Accepted: 04/23/2020] [Indexed: 02/01/2023]
Abstract
Myelomeningocele (MMC) is a spinal cord congenital defect that leads to paraplegia, bladder incontinence and bowel dysfunction. A randomized human trial demonstrated that in utero surgical repair of the MMC defect improves lower limb motor function. However, functional recovery remains incomplete. Stem cell therapy has recently generated great interest in the field of prenatal repair of MMC. In this systematic review we attempt to provide an overview of the current application of stem cells in different animal models of MMC. Publications were retrieved from PubMed and Cochrane Library databases. This process yielded twenty-two studies for inclusion in this review, experimenting five different types of stem cells: human embryonic stem cells, neural stem cells, induced pluripotent stem cells, human amniotic fluid stem cells, and mesenchymal stem cells (MSCs). Rodents and ovine were the two major species used for animal model studies. The source, the aims, and the main results were analyzed. Stem cell therapy appears to be a promising candidate for prenatal repair of MMC, especially MSCs. Further explorations in ovine and rodent models, reporting clinical and functional results, are necessary before an application in humans.
Collapse
|
22
|
Ojeda-Hernández DD, Canales-Aguirre AA, Matias-Guiu J, Gomez-Pinedo U, Mateos-Díaz JC. Potential of Chitosan and Its Derivatives for Biomedical Applications in the Central Nervous System. Front Bioeng Biotechnol 2020; 8:389. [PMID: 32432095 PMCID: PMC7214799 DOI: 10.3389/fbioe.2020.00389] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/07/2020] [Indexed: 12/12/2022] Open
Abstract
It is well known that the central nervous system (CNS) has a limited regenerative capacity and that many therapeutic molecules cannot cross the blood brain barrier (BBB). The use of biomaterials has emerged as an alternative to overcome these limitations. For many years, biomedical applications of chitosan have been studied due to its remarkable biological properties, biocompatibility, and high versatility. Moreover, the interest in this biomaterial for CNS biomedical implementation has increased because of its ability to cross the BBB, mucoadhesiveness, and hydrogel formation capacity. Several chitosan-based biomaterials have been applied with promising results as drug, cell and gene delivery vehicles. Moreover, their capacity to form porous scaffolds and to bear cells and biomolecules has offered a way to achieve neural regeneration. Therefore, this review aims to bring together recent works that highlight the potential of chitosan and its derivatives as adequate biomaterials for applications directed toward the CNS. First, an overview of chitosan and its derivatives is provided with an emphasis on the properties that favor different applications. Second, a compilation of works that employ chitosan-based biomaterials for drug delivery, gene therapy, tissue engineering, and regenerative medicine in the CNS is presented. Finally, the most interesting trends and future perspectives of chitosan and its derivatives applications in the CNS are shown.
Collapse
Affiliation(s)
- Doddy Denise Ojeda-Hernández
- Biotecnología Industrial, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Zapopan, Mexico
| | - Alejandro A Canales-Aguirre
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Jorge Matias-Guiu
- Servicio de Neurología, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - Ulises Gomez-Pinedo
- Servicio de Neurología, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - Juan C Mateos-Díaz
- Biotecnología Industrial, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Zapopan, Mexico
| |
Collapse
|
23
|
Jin C, Zhu Y, Wu Y, Tang Z, Tao L, Wei Y. RAI3 knockdown enhances osteogenic differentiation of bone marrow mesenchymal stem cells via STAT3 signaling pathway. Biochem Biophys Res Commun 2020; 524:516-522. [PMID: 32014253 DOI: 10.1016/j.bbrc.2020.01.098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 01/16/2020] [Indexed: 01/23/2023]
Abstract
Bone marrow mesenchymal stem cells (BMSCs), which have multipotential differentiation and self-renewal ability, have been becoming an attractive source of seed cells for bone tissue engineering. Nonetheless, the precise underlying mechanisms of osteogenesis of BMSCs have not been fully understood. Retinoic acid-induced gene 3 (RAI3) has been found to play important roles in mesenchymal stem cells (MSCs) adipogenesis in our previous study. However, its function in the osteogenic differentiation of BMSCs remains unknown. In this study, we found that RAI3 was significantly reduced in osteogenically differentiated BMSCs; RAI3 knockdown promoted osteogenesis of BMSCs both in vitro and in vivo. Moreover, we found RAI3 knockdown significantly upregulated the expression level of phosphorylated signal transducer and activator of transcription 3 (p-STAT3), and AG-490 which can inhibit the STAT3 signaling reversed the enhancing effect of RAI3 knockdown on the osteogenic differentiation of BMSCs. These results suggest that RAI3 plays important roles in BMSCs osteogenesis with an involvement of the STAT3 signaling, which might open a new avenue to explore BMSCs osteogenesis for the application of BMSCs in bone regeneration.
Collapse
Affiliation(s)
- Chanyuan Jin
- The Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, PR China; The Second Clinical Division of Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Yedan Zhu
- The Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, PR China; The Second Clinical Division of Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Yuwei Wu
- The Second Clinical Division of Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Zhihui Tang
- The Second Clinical Division of Peking University School and Hospital of Stomatology, Beijing, PR China.
| | - Lei Tao
- The Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, PR China.
| | - Yen Wei
- The Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, PR China.
| |
Collapse
|
24
|
Wei X, Cao S, Ma W, Zhang C, Gu H, Liu D, Luo W, Bai Y, Wang W, Yuan Z. Intra-Amniotic Delivery of CRMP4 siRNA Improves Mesenchymal Stem Cell Therapy in a Rat Spina Bifida Model. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 20:502-517. [PMID: 32330869 PMCID: PMC7177192 DOI: 10.1016/j.omtn.2020.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 03/13/2020] [Indexed: 11/16/2022]
Abstract
Neural tube defects (NTDs) result in prenatal mortality and lifelong morbidity, and available treatments have limited efficacy. We previously suggested that prenatal bone marrow-derived mesenchymal stem cell (BMSC) transplantation could treat neuron deficiency in NTD rats; however, BMSC-based therapy is limited by the low survival rate of BMSCs when used to treat severe NTDs. Herein, a new therapy using combined BMSC transplantation and small interfering RNA of collapsin response mediator protein 4 (CRMP4 siRNA), which was identified as a novel potential target for the NTD treatment, is proposed. The intra-amniotic CRMP4 siRNA, BMSC, and CRMP4 siRNA + BMSC injections repaired skin lesions, improved motor neural function, reduced neuronal apoptosis, and promoted expression of neural differentiation-related molecules and neurotrophic factors in the spinal cord of spina bifida rat fetuses. Therapeutic effects in the CRMP4 siRNA + BMSC injection group were superior to those of the CRMP4 siRNA only or BMSC only injection groups. CRMP4 siRNA + BMSC injection resulted in a 45.38% reduction in the skin lesion area and significantly shorter latency and higher amplitude of motor-evoked potentials (MEPs) in spina bifida fetuses. Our results suggest that intrauterine Ad-CRMP4 siRNA delivery with BMSCs is an innovative platform for developing fetal therapeutics to safely and efficaciously treat NTDs.
Collapse
Affiliation(s)
- Xiaowei Wei
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, P.R. China
| | - Songying Cao
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, P.R. China.
| | - Wei Ma
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, P.R. China
| | - Chaonan Zhang
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, P.R. China
| | - Hui Gu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, P.R. China
| | - Dan Liu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, P.R. China
| | - Wenting Luo
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, P.R. China
| | - Yuzuo Bai
- Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, P.R. China
| | - Weilin Wang
- Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, P.R. China
| | - Zhengwei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, P.R. China.
| |
Collapse
|
25
|
Shkarin R, Shkarin A, Shkarina S, Cecilia A, Surmenev RA, Surmeneva MA, Weinhardt V, Baumbach T, Mikut R. Quanfima: An open source Python package for automated fiber analysis of biomaterials. PLoS One 2019; 14:e0215137. [PMID: 30973910 PMCID: PMC6459545 DOI: 10.1371/journal.pone.0215137] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 03/27/2019] [Indexed: 12/19/2022] Open
Abstract
Hybrid 3D scaffolds composed of different biomaterials with fibrous structure or enriched with different inclusions (i.e., nano- and microparticles) have already demonstrated their positive effect on cell integration and regeneration. The analysis of fibers in hybrid biomaterials, especially in a 3D space is often difficult due to their various diameters (from micro to nanoscale) and compositions. Though biomaterials processing workflows are implemented, there are no software tools for fiber analysis that can be easily integrated into such workflows. Due to the demand for reproducible science with Jupyter notebooks and the broad use of the Python programming language, we have developed the new Python package quanfima offering a complete analysis of hybrid biomaterials, that include the determination of fiber orientation, fiber and/or particle diameter and porosity. Here, we evaluate the provided tensor-based approach on a range of generated datasets under various noise conditions. Also, we show its application to the X-ray tomography datasets of polycaprolactone fibrous scaffolds pure and containing silicate-substituted hydroxyapatite microparticles, hydrogels enriched with bioglass contained strontium and alpha-tricalcium phosphate microparticles for bone tissue engineering and porous cryogel 3D scaffold for pancreatic cell culturing. The results obtained with the help of the developed package demonstrated high accuracy and performance of orientation, fibers and microparticles diameter and porosity analysis.
Collapse
Affiliation(s)
- Roman Shkarin
- Laboratory for Applications of Synchrotron Radiation, Karlsruhe Institute of Technology, Karlsruhe, Germany
- Institute for Automation and Applied Computer Science, Karlsruhe Institute of Technology, Karlsruhe, Germany
- * E-mail:
| | - Andrei Shkarin
- Laboratory for Applications of Synchrotron Radiation, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Svetlana Shkarina
- Physical Materials Science and Composite Materials Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, Russia
- Institute for Photon Science and Synchrotron Radiation, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Angelica Cecilia
- Institute for Photon Science and Synchrotron Radiation, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Roman A. Surmenev
- Physical Materials Science and Composite Materials Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, Russia
| | - Maria A. Surmeneva
- Physical Materials Science and Composite Materials Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, Russia
| | - Venera Weinhardt
- Laboratory for Applications of Synchrotron Radiation, Karlsruhe Institute of Technology, Karlsruhe, Germany
- Institute for Photon Science and Synchrotron Radiation, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
- Centre for Organismal Studies, COS, Heidelberg University, Heidelberg, Germany
| | - Tilo Baumbach
- Laboratory for Applications of Synchrotron Radiation, Karlsruhe Institute of Technology, Karlsruhe, Germany
- Institute for Photon Science and Synchrotron Radiation, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Ralf Mikut
- Institute for Automation and Applied Computer Science, Karlsruhe Institute of Technology, Karlsruhe, Germany
| |
Collapse
|
26
|
Zhang WL, Chi CT, Meng XH, Liang SD. miRNA‑15a‑5p facilitates the bone marrow stem cell apoptosis of femoral head necrosis through the Wnt/β‑catenin/PPARγ signaling pathway. Mol Med Rep 2019; 19:4779-4787. [PMID: 30957181 PMCID: PMC6522831 DOI: 10.3892/mmr.2019.10130] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 03/19/2019] [Indexed: 12/14/2022] Open
Abstract
Bone marrow stem cells (BMSCs) are a group cells that function as an underlying cell source for bone tissue regeneration. However, the molecular mechanisms of how BMSCs are induced into apoptosis remains unclear. In the present study, it was demonstrated that the molecular mechanisms of BMSCs were exerted via microRNA-15a-5p (miR-15a-5p) in femoral head necrosis (FHN). Briefly, miRNA-15a-5p expression was elevated in a rat model of FHN. Overexpression of miR-15a-5p promoted the apoptosis of BMSCs and reduced cell growth through the Wnt/β-catenin/peroxisome proliferator-activated receptor γ (PPARγ) signaling pathway. Downregulation of miR-15a-5p reduced the apoptosis of BMSCs and promoted cell growth through the Wnt/β-catenin/PPARγ signaling pathway. The activation of Wnt attenuated the effects of miR-15a-5p on the apoptosis of BMSCs via the β-catenin/PPARγ signaling pathway. In conclusion, the present results indicated that miRNA-15a-5p was involved in the regulation of the apoptosis of BMSCs through regulating the Wnt/β-catenin/PPARγ signaling pathway, which may serve an important role in the regulation of FHN.
Collapse
Affiliation(s)
- Wan-Li Zhang
- Department of Neurosurgery, Hongqi Hospital Affiliated with Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Cheng-Tao Chi
- Department of Neurosurgery, Hongqi Hospital Affiliated with Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Xiang-Hui Meng
- Department of Neurosurgery, Hongqi Hospital Affiliated with Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Shao-Dong Liang
- Department of Neurosurgery, Hongqi Hospital Affiliated with Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| |
Collapse
|
27
|
Liem NT, Chinh VD, Thinh NT, Minh ND, Duc HM. Improved Bowel Function in Patients with Spina Bifida After Bone Marrow-Derived Mononuclear Cell Transplantation: A Report of 2 Cases. AMERICAN JOURNAL OF CASE REPORTS 2018; 19:1010-1018. [PMID: 30143601 PMCID: PMC6120349 DOI: 10.12659/ajcr.909801] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/21/2018] [Indexed: 11/09/2022]
Abstract
BACKGROUND Bowel dysfunction is observed in 42.2-71.2% of patients with spina bifida. Traditional treatments yield limited results. The objective of this paper is to report on improvement in bowel function in 2 children with spina bifida following bone marrow-derived mononuclear cells transplantation. CASE REPORT Two patients - 14 years old and 11 years old - with bowel dysfunction after myelomeningocele repair underwent 2 BMMNC transplantations without complications. Those patients had normal defecation, assessed through follow-ups of 21 months and 16 months, respectively. CONCLUSIONS BMMNC transplantation can improve bowel function, as demonstrated in 2 patients with spina bifida.
Collapse
Affiliation(s)
- Nguyen Thanh Liem
- Stem Cell and Immune Cell Department, Vinmec Research Institute of Stem Cell and Gene Technology, Times City, Ha Noi, Vietnam
| | - Vu Duy Chinh
- Center for Excellence for Autism and Cerebral Palsy, Vinmec International Hospital, Times City, Ha Noi, Vietnam
| | - Nguyen Thi Thinh
- Pediatric Department, Vinmec International Hospital, Times City, Ha Noi, Vietnam
| | - Ngo Duy Minh
- Pediatric Department, Vinmec International Hospital, Times City, Ha Noi, Vietnam
| | - Hoang Minh Duc
- Stem Cell and Immune Cell Department, Vinmec Research Institute of Stem Cell and Gene Technology, Times City, Ha Noi, Vietnam
| |
Collapse
|
28
|
Zhang J, Chen Y, Xu J, Wang J, Li C, Wang L. Tissue engineering using 3D printed nano-bioactive glass loaded with NELL1 gene for repairing alveolar bone defects. Regen Biomater 2018; 5:213-220. [PMID: 30094060 PMCID: PMC6077810 DOI: 10.1093/rb/rby015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/14/2018] [Accepted: 05/21/2018] [Indexed: 12/22/2022] Open
Abstract
The purposes of this study were to construct a novel tissue engineered bone composed of 3D-printed bioactive glass block/chitosan nanoparticles (BD/CSn) composites loaded with Nel-like Type I molecular-1 DNA (pDNA-NELL1) and/or bone marrow mesenchymal stem cells (BMSCs), and study their osteogenic activities by repairing bone defects in rhesus monkeys. CSn with NELL1 gene plasmid and rhesus monkey BMSCs were composited with a BD scaffold to prepare the tissue-engineered bone. Four adult female rhesus monkeys with 10- to 12-years old and 5-7 kg in weight were used in animal experiments. The first and second premolar teeth from four regions of each monkey were removed to form bone defects with size of 10 × 10 × 5 mm, which were then implanted with above-mentioned tissue engineered bone. At 12 weeks after the implantation, gross observations, X-ray and micro-CT observations revealed that the new bone was extremely close to normal bone in mass, density, hardness, and structure. The bony cortex was smooth and closely connected to the surrounding normal bone. Histological observations revealed moderate inflammation in the repair area, and the new bone tissues were similar to normal ones. In conclusion, tissue engineered bone of this study exhibited good osteoconductivity for promoting the formation of new alveolar bone tissue, and NELL1 gene played a promotional role in bone regeneration.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Periodontology, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China.,Department of Stomatology, Foshan Woman and Children's Hospital, Foshan, Guangdong, China
| | - Yang Chen
- Department of Spinal Surgery, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Jing Xu
- Department of Stomatology, Jiangyin People's Hospital of Southeast University, Jiangyin, Jiangsu, China
| | - Jingjing Wang
- Department of Stomatology, Foshan Woman and Children's Hospital, Foshan, Guangdong, China
| | - Chengzhang Li
- Department of Periodontology, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Liyan Wang
- Department of Stomatology, Foshan Woman and Children's Hospital, Foshan, Guangdong, China
| |
Collapse
|
29
|
Silva MDA, Leite YKDC, de Carvalho CES, Feitosa MLT, Alves MMDM, Carvalho FADA, Neto BCV, Miglino MA, Jozala AF, de Carvalho MAM. Behavior and biocompatibility of rabbit bone marrow mesenchymal stem cells with bacterial cellulose membrane. PeerJ 2018; 6:e4656. [PMID: 29736332 PMCID: PMC5933324 DOI: 10.7717/peerj.4656] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 04/01/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Tissue engineering has been shown to exhibit great potential for the creation of biomaterials capable of developing into functional tissues. Cellular expansion and integration depends on the quality and surface-determinant factors of the scaffold, which are required for successful biological implants. The objective of this research was to characterize and evaluate the in vitro characteristics of rabbit bone marrow mesenchymal stem cells (BM-MSCs) associated with a bacterial cellulose membrane (BCM). We assessed the adhesion, expansion, and integration of the biomaterial as well as its ability to induce macrophage activation. Finally, we evaluated the cytotoxicity and toxicity of the BCM. METHODS Samples of rabbit bone marrow were collected. Mesenchymal stem cells were isolated from medullary aspirates to establish fibroblast colony-forming unit assay. Osteogenic, chondrogenic, and adipogenic differentiation was performed. Integration with the BCM was assessed by scanning electron microscopy at 1, 7, and 14 days. Cytotoxicity was assessed via the production of nitric oxide, and BCM toxicity was assessed with the MTT assay; phagocytic activity was also determined. RESULTS The fibroblastoid colony-forming unit (CFU-F) assay showed cells with a fibroblastoid morphology organized into colonies, and distributed across the culture area surface. In the growth curve, two distinct phases, lag and log phase, were observed at 15 days. Multipotentiality of the cells was evident after induction of osteogenic, chondrogenic, and adipogenic lineages. Regarding the BM-MSCs' bioelectrical integration with the BCM, BM-MSCs were anchored in the BCM in the first 24 h. On day 7 of culture, the cytoplasm was scattered, and on day 14, the cells were fully integrated with the biomaterial. We also observed significant macrophage activation; analysis of the MTT assay and the concentration of nitric oxide revealed no cytotoxicity of the biomaterial. CONCLUSION The BCM allowed the expansion and biointegration of bone marrow progenitor cells with a stable cytotoxic profile, thus presenting itself as a biomaterial with potential for tissue engineering.
Collapse
Affiliation(s)
- Marcello de Alencar Silva
- Integrated Nucleus of Morphology and Stem Cell Research, Federal University of Piauí, Teresina, Piauí, Brazil
| | | | | | - Matheus Levi Tajra Feitosa
- Integrated Nucleus of Morphology and Stem Cell Research, Federal University of Piauí, Teresina, Piauí, Brazil
| | | | | | - Bartolomeu Cruz Viana Neto
- Department of Physics/Advanced Microscopy Multiuser Laboratory/Laboratory of Physics Material, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Maria Angélica Miglino
- Departament of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Angela Faustino Jozala
- Laboratory of Industrial Microbiology and Fermentation Process, University of Sorocaba, Sorocaba, São Paulo, Brazil
| | | |
Collapse
|
30
|
Wu SP, Yang Z, Li FR, Liu XD, Chen HT, Su DN. Smad7-overexpressing rat BMSCs inhibit the fibrosis of hepatic stellate cells by regulating the TGF-β1/Smad signaling pathway. Exp Ther Med 2017; 14:2568-2576. [PMID: 28962196 PMCID: PMC5609222 DOI: 10.3892/etm.2017.4836] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 04/28/2017] [Indexed: 11/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are able to differentiate into hepatocytes, promote the regeneration of hepatic cells and inhibit the progression of hepatic fibrosis. Transforming growth factor (TGF)-β1 is one of the key factors in the development of liver fibrosis, which also promotes extracellular matrix (ECM) formation. Drosophila mothers against decapentaplegic 7 (Smad7) is an essential negative regulator in the TGF-β1/Smad signaling pathway. In the present study, bone mesenchymal stem cells (BMSCs) were isolated from rat bone marrow and transfected with lentiviral vectors carrying the Smad7 gene. Smad7-enhanced green fluorescent protein (EGFP)-BMSCs stably expressing Smad7 were subsequently co-cultured with hepatic stellate cells (HSCs) for 48 h. Smad7 and TGF-β1 levels in the culture medium were detected using ELISA, and the levels of collagen (Col) I, Col III, laminin (LN) and hyaluronic acid (HA) were measured using immunoassays. The early apoptosis rates of HSCs were determined via flow cytometry. Reverse transcription-quantitative polymerase chain reaction and western blotting were performed to evaluate the mRNA and protein expression profiles, respectively. The results indicated that Smad7-EGFP-BMSCs stably expressing Smad7 were successfully constructed. Upon co-culturing with rat Smad7-EGFP-BMSCs, the early apoptotic rate of HSCs was significantly increased (P<0.05). Levels of Smad7 in the culture medium were also significantly increased (P<0.05), whereas the levels of TGF-β1, Col I, Col III, LN and HA were significantly decreased (P<0.05). Furthermore, the mRNA and protein levels of Smad7 and matrix metalloproteinase 1 were significantly increased (P<0.05), whereas those of TGF-β1, α-SMA, Smad2, smad3, TGF-β receptor I, Col I, tissue inhibitors of metalloproteinase-1 and Col III were significantly decreased. The results of the present study suggest that rat BMSCs overexpressing Smad7 may inhibit the fibrosis of HSCs by regulating the TGF-β1/Smad signaling pathway. This provides a novel insight into future treatments for liver fibrosis.
Collapse
Affiliation(s)
- Shi-Pin Wu
- Department of Infectious Diseases, The Second Clinical Medical College, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Zhi Yang
- Department of Infectious Diseases, The Second Clinical Medical College, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Fu-Rong Li
- The Key Laboratory of Stem Cell and Cellular Therapy, The Second Clinical Medical College, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Xiao-Di Liu
- Department of Infectious Diseases, The Second Clinical Medical College, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Hong-Tao Chen
- Department of Infectious Diseases, The Second Clinical Medical College, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Dong-Na Su
- Department of Infectious Diseases, The Second Clinical Medical College, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong 518020, P.R. China
| |
Collapse
|
31
|
Ruan SQ, Yan L, Deng J, Huang WL, Jiang DM. Preparation of a biphase composite scaffold and its application in tissue engineering for femoral osteochondral defects in rabbits. INTERNATIONAL ORTHOPAEDICS 2017; 41:1899-1908. [DOI: 10.1007/s00264-017-3522-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 05/15/2017] [Indexed: 12/30/2022]
|
32
|
Xun Q, Wang H, He F. Bone marrow mesenchymal stem cells for treatment of pancreatic diseases: Research status and prospects. Shijie Huaren Xiaohua Zazhi 2016; 24:3232-3237. [DOI: 10.11569/wcjd.v24.i21.3232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The incidence of pancreatic diseases is increasing year by year. Current treatments for pancreatic diseases are mainly symptomatic, and the research on the repair and restoration of function of the pancreatic cells progresses slowly. Stem cells have been widely used in the treatment of diseases in recent years because of their ability of multi-directional differentiation and repair of cell damage caused by disease and injury. Numerous studies confirm that pancreatic stem cells after transplantation can differentiate into pancreatic cells and play an important role in the recovery of external secretory function and repair of the damaged pancreatic cells. Particularly, both in vivo and in vitro studies show that bone marrow mesenchymal stem cells have achieved remarkable results in the treatment of pancreatic diseases, laying a theoretical and practical basis for clinical treatment of pancreatic diseases with stem cells. This article outlines the progress in treatment of acute pancreatitis, chronic pancreatitis and pancreatic cancer with bone marrow mesenchymal stem cells, demonstrating that stem cells are expected to become one of new methods for the treatment of pancreatic diseases.
Collapse
|